WO2009077483A1 - Antibodies against human nkg2d and uses thereof - Google Patents

Antibodies against human nkg2d and uses thereof Download PDF

Info

Publication number
WO2009077483A1
WO2009077483A1 PCT/EP2008/067499 EP2008067499W WO2009077483A1 WO 2009077483 A1 WO2009077483 A1 WO 2009077483A1 EP 2008067499 W EP2008067499 W EP 2008067499W WO 2009077483 A1 WO2009077483 A1 WO 2009077483A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
hnkg2d
nkg2d
seq
antibodies
Prior art date
Application number
PCT/EP2008/067499
Other languages
English (en)
French (fr)
Inventor
Birgitte URSØ
Peter Andreas Nicolai Reumert Wagtmann
Inger Lund Pedersen
Anders Svensson
Original Assignee
Novo Nordisk A/S
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=40336450&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2009077483(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to CN200880127171.3A priority Critical patent/CN101945893B/zh
Priority to AU2008337517A priority patent/AU2008337517B2/en
Priority to ES08861706.3T priority patent/ES2483942T5/es
Priority to US12/747,095 priority patent/US9127064B2/en
Priority to BRPI0821658A priority patent/BRPI0821658B8/pt
Priority to JP2010537469A priority patent/JP5591712B2/ja
Priority to EP08861706.3A priority patent/EP2222706B2/en
Priority to PL08861706T priority patent/PL2222706T5/pl
Priority to CA2708854A priority patent/CA2708854C/en
Priority to DK08861706.3T priority patent/DK2222706T4/en
Application filed by Novo Nordisk A/S filed Critical Novo Nordisk A/S
Priority to RU2010125034/10A priority patent/RU2563343C2/ru
Publication of WO2009077483A1 publication Critical patent/WO2009077483A1/en
Priority to US12/605,187 priority patent/US7879985B2/en
Priority to IL205866A priority patent/IL205866A/en
Priority to ZA2010/03759A priority patent/ZA201003759B/en
Priority to US14/809,680 priority patent/US20160024214A1/en
Priority to US15/707,393 priority patent/US10526409B2/en
Priority to US16/655,646 priority patent/US20200031939A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2299/00Coordinates from 3D structures of peptides, e.g. proteins or enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/74Inducing cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to antibodies against human NKG2D (hNKG2D) and their use in treating or preventing diseases and disorders in human patients.
  • hNKG2D human NKG2D
  • the immunoreceptor NKG2D is normally expressed on human CD8 + T cells and NK cells.
  • the human NKG2D (hNKG2D) homodimeric receptor functions as a co-stimulator of TCR and CD28+TCR signalling via its DAP10 association, whereas in NK cells it functions as a direct activator.
  • Various ligands for hNKG2D have been identified and characterized, including the MHC Class l-related ligands MICA and MICB, the UL16-binding protein (ULBP) family, and the retinoic acid early transcript-1 (RAET1 ) family.
  • hNKG2D In chronic autoimmune diseases such as rheumatoid arthritis, hNKG2D is expressed on a sub-set of CD4 + CD28 " T cells and is involved in stimulation of their proliferation and IFN ⁇ production, and MIC expression is upregulated (Groh et al. , PNAS 2003; 100:9452). It has also been shown that CD4+ hNKG2D-expressing T cells in Crohn's disease mediate inflammatory and cytotoxic responses through MICA interactions (Allez et a/. , Gastroenterology 2007; 132:2346-2358).
  • NKG2D is an essential driver in autoimmune inflammation
  • NOD mice a murine model of diabetes (NOD mice) by a monoclonal antibody (mAb) binding to and blocking murine NKG2D (CX5) (Ogasawara et a/., Immunity 2004; 20:757-767), suggesting therapeutic applications for anti-NKG2D antibodies.
  • mAb monoclonal antibody
  • CX5 murine NKG2D
  • Such applications have been described in, e.g., US20050158307, WO2005097160, WO2005115517, and WO2006024367.
  • the present invention provides isolated anti-hNKG2D monoclonal antibodies useful for therapeutic applications in humans.
  • the antibodies are fully human or humanized to minimize the risk for immune responses against the antibodies when administered to a patient.
  • other antigen-binding molecules such as, e.g., antigen- binding antibody fragments, antibody derivatives, and multi-specific molecules, can be designed or derived from such antibodies.
  • the antibodies are characterized by one or more functional properties, or by a combination of functional properties.
  • Exemplary properties include, e.g., preventing hNKG2D-mediated activation of hNKG2D-expressing NK or T cell; competing with at least one natural hNKG2D ligand, or with several ligands, in binding to hNKG2D; reducing the amount of hNKG2D on the surface of a hNKG2D-expressing NK or T cell; binding also cy- nomolgous and/or rhesus NKG2D; binding only one antibody molecule per hNKG2D dimer; cross-linking no more than 2 hNKG2D dimers when added to hNKG2D-expressing NK and/or T cells; having insignificant agonist effect on hNKG2D signalling upon binding; and/or binding to hNKG2D with a dissociation constant (KD) of 1 nM or less.
  • KD dissociation constant
  • Certain anti-hNKG2D antibodies of the invention may also or alternatively compete with, bind to essentially the same epi- tope as, or bind with the same or higher affinity as, one or more particular human anti- hNKG2D antibodies described herein, including antibodies MS and 21 F2.
  • the antibodies are also or alternatively more capable of competing with or blocking hNKG2D-binding of MS and/or 21 F2 than known murine anti-hNKG2D antibodies (e.g., the ones described above).
  • the antibodies bind to the same hNKG2D epitope as MS and/or 21 F2.
  • the antibodies also or alternatively bind the same epitope as MS.
  • the antibodies also or alternatively bind the same epitope as 21 F2.
  • antibodies provided by and/or used in embodiments of this invention may exhibit three, four, or more of the above-referenced features.
  • the antibodies also or alternatively comprise one or more paratopes and/or antigen-binding sequences that are identical or similar to MS or 21 F2 paratopes and/or antigen-binding sequences described herein.
  • the invention provides for nucleic acids encoding antibodies of the invention, expression vectors comprising such nucleic acids, host cells comprising such nu- cleic acids, host cells producing antibodies of the invention, and methods of producing anti- hNKG2D antibodies by culturing such host cells under appropriate conditions.
  • Antibody-binding fragments of such antibodies as well as molecules comprising such antigen-binding fragments, including engineered antibody fragments, antibody deriva- tives, bispecific antibodies and other multispecific molecules, are also provided.
  • compositions and kits or other articles that comprise such antibodies or other molecules also are provided.
  • autoimmune and/or inflammatory diseases or disorders including, but not limited to rheumatoid arthritis, inflammatory bowel disease (IBD) including Crohn's disease and ulcerative colitis, systemic erythromatosis lupus (SLE), psoriasis, psoriatic arthritis, multiple sclerosis, celiac disease, viral disease (such as, e.g., viral hepatitis), and transplant rejection of various organs and tissues (including, but not limited to, heart and bone marrow), using such antibodies, molecules, and compositions.
  • IBD inflammatory bowel disease
  • SLE systemic erythromatosis lupus
  • psoriasis psoriatic arthritis
  • multiple sclerosis celiac disease
  • viral disease such as, e.g., viral hepatitis
  • transplant rejection of various organs and tissues including, but not limited to, heart and bone marrow
  • Figure 1 shows analyses of exemplary sera from hNKG2D-immunized mice from the KM mouseTM strain.
  • Figure 2 shows an example of a human antibody in the form of a hybridoma supernatant bound specifically to NKG2D expressing cells (A) but not to the same cell-line not expressing NKG2D (B). Antibody was added to the cells in the form of hybridoma supernatant. The binding of a directly labelled positive control, murine anti-NKG2D antibody 149810, to NKG2D expressing cells (C) and non expressing cells (D), is also shown. The black outline represents background staining, and solid peaks represent specific staining.
  • Figure 3 demonstrates dose-response of NKG2D-binding to NKG2D-expressing cells of recombinantly expressed and purified fully human lgG4 antibodies (16F16, 16F31 , MS, and 21 F2) as compared to commercial murine antibodies (ON72 and 149810).
  • Figure 4 shows the amino acid sequences for the heavy (H) and light (L) chains of human anti-hNKG2D antibodies 16F16, and 16F31 (A), and MS and 21 F2 (B) of lgG4 iso- type, highlighting variable regions (bold) and CDR regions (underlined).
  • the corresponding sequence identifiers for the amino acid sequences and the various highlighted portions are provided in Table 1.
  • Figure 5 shows alignments of VH and VL sequences with the corresponding recom- bined germline sequences. CDR regions are indicated by bold Kabat numbers and somatic hypermutations are indicated by bold underlined text.
  • A 16F16 lgG4 H chain;
  • B 16F16 lgG4 L chain;
  • C 16F31 lgG4 H chain;
  • D 16F31 lgG4 L chain;
  • E MS lgG4 H chain;
  • F MS lgG4 L chain;
  • G 21 F2 lgG4 H chain;
  • H 21 F2 lgG4 L chain.
  • SEQ ID NOS:27-30 correspond to recombined VH3_21/D3-9/JH4, VKI_L15/JK2, VH3_20/D3-10/JH6, and VKIII_A27/JK3, respectively, and SEQ ID NOS 60-63 correspond to recombined VH4_59//JH3, VKIII_A27/JK1 , VH5_51/D3_10_R3/JH4, and VKIII_L6/JK1 , respectively.
  • Figure 6 shows blockade of ligand- (MICA-) binding by an exemplary human anti- NKG2D antibody, demonstrated by blockade of ligand binding by preincubation with antibody in a hybridoma supernatant.
  • the outline represents background, grey represents ligand binding without pre-incubation, and black with dotted line represents ligand binding with pre- incubation.
  • Figure 7 shows a dose-response curve obtained when analyzing various concentrations of recombinantly expressed and purified fully human anti-hNKG2D antibodies (16F16, 16F31 , MS, and 21 F2; lgG4 isotype), giving the IC50 and dose needed for full blockade of 1 ⁇ g MICA-mFc binding.
  • Figure 8 shows that NKG2D-binding of ON72 to NKG2D was completely prevented by pre-incubation with hybridoma supernatant containing 16F16. Outline represents background, gray represents ON72-binding without pre-incubation, and black dotted represents ON72-binding with pre-incubation.
  • Figure 9 shows the capability of fully human anti-hNKG2D antibodies to block the subsequent binding of murine anti-hNKG2D antibodies to NKG2D, or vice versa.
  • 16F16 Pre-incubation with recombinantly expressed and purified 16F16 (0.3 ⁇ g; lgG4 isotype) prevented ON72 (0.3 ⁇ g) from binding to NKG2D.
  • Antibody 149810 only demonstrated approximately 50% cross-inhibition of recombinantly expressed and purified 16F16 (lgG4 isotype), when tested at 1 : 1 (0.3 ⁇ g, 0.3 ⁇ g) and at 3: 1 (1 ⁇ g, 0.3 ⁇ g) of antibody concentration, 149810 to 16F16, respectively, again likely showing differences in the binding epitope on NKG2D.
  • Figure 10 shows staining of rhesus or cynomologous (cyno) cells with ON72 and 16F16 antibody purified from original hybridoma.
  • A cyno NK cells
  • B cyno CD8+ T cells
  • C rhesus NK cells
  • D rhesus CD8+ T cells.
  • MFI mean fluorescent intensity
  • Figure 1 1 shows the binding of human antibody MS to human or cynomolgous CD8- positive cells in perifieral blood mononuclear cells (PBMCs) at different antibody concentrations, demonstrating that the affinity to human and cynomologous NKG2D is similar.
  • PBMCs perifieral blood mononuclear cells
  • Figure 12 shows that addition of ligand-blocking antibodies, (ON72 or recombinantly expressed and purified 16F16 (lgG4 isotype)), blocked NK-mediated killing of MICA- expressing target cells in a dose-dependent fashion in a 51 Cr-release assay.
  • Figure 13 shows that recombinantly expressed and purified 16F16 and 16F31 (both lgG4 isotype) were capable of inhibiting killing of both MICA (A) and ULBP3 (B) bearing target cells (BaF/3) by NK-92 cells in a dose dependent manner, with near total blockade by 16F16 at 0.8 ⁇ g/ml for both ligands, and partial blockade by 16F31 at the highest tested dose of 20 ⁇ g/ml for both ligands.
  • Figure 14 shows that recombinantly expressed and purified MS, 21 F2, and 16F16 (all lgG4 isotype) were capable of inhibiting NK-mediated killing of ligand-expressing target cells.
  • A inhibition of NK-92 cells killing of ULBP3-BaF/3 cells by MS or 21 F2.
  • B inhibition of NKL cells killing BaF/3-MICA cells by MS or 16F16.
  • Figure 15 shows antibody-induced reduction of cell-surface NKG2D, using BaF/3 cells transfected with NKG2D and DAP10.
  • Figure 16 shows MS antibody-induced reduction of cell-surface NKG2D, using BaF/3 cells transfected with NKG2D and DAP10 (performed as for figure 15) (A), or freshly prepared human NK cells from peripheral blood (B).
  • A the human NK cells were incu- bated overnight in the presence of human serum, to mimic a situation in blood with IgGs present, and varying concentrations of MS antibody. Maximum downmodulation was achieved even at the lowest concentration, corresponding to about 60% receptor saturation measured in binding assay under similar conditions on NKG2D+ NK cells.
  • Figure 17 shows the percentage reduction of cell-surface NKG2D on human NK cells after over-night incubation with indicated 21 F2 antibody concentrations.
  • Figure 18 shows the effect of ON72, MS, and 21 F2 on surface-presented NKG2D in different types of cells in human blood samples, at the indicated time points.
  • the concentration of each antibody was 0.1 ⁇ g/ml. While not being limited to theory, the reduction of surface-presented NKG2D in the experiments likely represents NKG2D internalization.
  • Figures (A) to (C) shows antibody-induced NKG2D internalization of NKG2D-expressing NK cells
  • Figure 19 shows the results of an assay testing for an agonistic effect of immobilized MS and ON72 on T cell proliferation, using 2 different sub-optimal doses of CD3 to allow for co-stimulation.
  • [CD3] 0.1 ng/ml
  • [CD3] 0.3 ng/ml.
  • T cell proliferation was assessed by CFSE dilution in a PBMC population stimulated with immobilized antibody as indicated for 3 days followed by IL-2 stimulation for four days.
  • CD28 stimulation is included as a positive control of co-stimulation. No significant agonistic effect could be detected for MS, whereas ON72 had a low but significant effect on T cell proliferation.
  • Figure 20 depicts 3-dimensional superimposed representations of hNKG2D dimer complexed with Fab-fragment(s) of anti-NKG2D antibody (MS or hzON72) or with MICA ligand.
  • the hNKG2D homodimer ('NKG2D') is shown in a surface representation with one of the monomers in a darker color than the other.
  • the Fab fragments ('MS' and 'hzON72', respectively) are indicated in black tube style while the MICA ('MICA') is indicated in a light schematic secondary structure representation style.
  • Figure 21 shows the epitope residues in the sequence (SEQ ID NO:2) of each NKG2D monomer unit of a hNKG2D dimer for MS Fab (A), hzON72 Fab (B) and a MICA molecule (C) in the sequences (SEQ ID NO:2) of the two hNKG2D monomer units.
  • NKG2D residues within 4.0 A distance from the crystal structure ligand atoms were considered to be part of the binding epitope and are underlined. Doubly underlined residues were involved in hydrogen-binding to the ligand.
  • A Binding epitope for a single MS Fab on hNKG2D monomer units 1 and 2 in a hNKG2D dimer.
  • Crystallographic monomers N and C were combined in the NKG2D monomer unit 1 , and crystallographic monomers M and D were combined in the NKG2D monomer unit 2.
  • the Lys 150 side chain atom N ⁇ was only involved in hydrogen-binding in one of the two crystallographically independent complexes. See also Tables 9-12.
  • Trp 166 was involved in hydrogen-bonding in one of the crystallographically independent molecular complexes (one hzON72 Fab molecule in complex with one hNKG2D monomer) but the distance was too far for hydrogen-binding in the other.
  • Figure 22 shows the hNKG2D molecules in surface representations with one of the monomers slightly darker than the other.
  • MS/hzON72/MICA Fab atoms are colored in black and are shown for the MS Fab (A), 2 hzON72 Fabs (B) and, for MICA, (C) and (D).
  • the relative binding orienta- tion to NKG2D can differ. This is indicated in the figure which shows the two possible relative binding orientations of MICA in (C) and (D) to MS. See also Tables 9-12 and 14-15.
  • hNKG2D and, unless otherwise stated or contradicted by context, the terms “NKG2D,” also known as “NKG2-D,” “CD314,” “D12S2489E,” “KLRK1 ,” “killer cell lectin-like receptor subfamily K, member 1 ,” and “KLRK1 ,” refer to a human killer cell activating receptor gene, its mRNA (e.g. , NCBI RefSeq NM_007360; SEQ ID NO:1), and its gene product (NCBI RefSeq NP_031386; SEQ ID NO:2), or naturally occurring variants thereof.
  • mRNA e.g. , NCBI RefSeq NM_007360; SEQ ID NO:1
  • NCBI RefSeq NP_031386 SEQ ID NO:2
  • the ligand-binding form of the hNKG2D receptor is a homodimer (Li et al,, Nat Immunol 2001 ;2:443-451 ).
  • the hNKG2D receptor is typically presented at the surface in complex with DAP10 (Wu et al, J Exp Med 2000; 192: 1059 et seq.; NCBI Accession No. AAG29425, AAD50293) and has been suggested to also form higher order complexes.
  • hNKG2D Any activity attributed herein to hNKG2D, e.g., cell activation, antibody recognition, etc., can also be attributed to hNKG2D in the form of a complex or higher-order complexes with DAP10, and/or other components.
  • antibody herein is used in the broadest sense and specifically includes full-length monoclonal antibodies, polyclonal antibodies, and, unless otherwise stated or contradicted by context, antigen-binding fragments, antibody variants, and multispecific molecules thereof, so long as they exhibit the desired biological activity.
  • a full-length antibody is a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CH1 , CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • the VH and VL regions can be further subdivided into regions of hypervariabil- ity, termed complementarily determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarily determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following or- der: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • An "antigen-binding fragment" of an antibody is a molecule that comprises a portion of a full-length antibody which is capable of detectably binding to the antigen, typically comprising one or more portions of at least the VH region.
  • Antigen-binding fragments include multivalent molecules comprising one, two, three, or more antigen-binding portions of an an- tibody, and single-chain constructs wherein the VL and VH regions, or selected portions thereof, are joined by synthetic linkers or by recombinant methods to form a functional, antigen-binding molecule. While some antigen-binding fragments of an antibody can be obtained by actual fragmentation of a larger antibody molecule (e.g. , enzymatic cleavage), most are typically produced by recombinant techniques.
  • antibody derivative and “immunoconjugate” are used interchangeably herein to denote molecules comprising a full-length antibody or an antigen-binding fragment thereof, wherein one or more amino acids are chemically modified, e.g., by alkylation, PEGy- lation, acylation, ester formation or amide formation or the like, e.g., for linking the antibody to a second molecule.
  • modifications include PEGylation (e.g., cysteine- PEGylation), biotinylation, radiolabelling, and conjugation with a second agent (such as a cytotoxic agent),
  • a “multispecific molecule” comprises an antibody, or an antigen-binding fragment thereof, which is associated with or linked to at least one other functional molecule (e.g. another peptide or protein such as another antibody or ligand for a receptor) thereby forming a molecule that binds to at least two different binding sites or target molecules.
  • exemplary multispecific molecules include bi-specific antibodies and antibodies linked to soluble receptor fragments or ligands.
  • human antibody is intended to include antibodies having variable regions in which both the framework and CDR regions are derived from (i.e. , are identical or essentially identical to) human germline immunoglobulin sequences. Furthermore, if the antibody contains a constant region, the constant region also is "derived from" human germline immunoglobulin sequences.
  • the human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in viva). However, the term “human antibody”, as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • humanized antibody is a human/non-human chimeric antibody that contains a minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hyper- variable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit, or non-human primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit, or non-human primate having the desired specificity, affinity, and capacity.
  • FR residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • hu- manized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR residues are those of a human immunoglobulin sequence.
  • the humanized antibody can optionally also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • hypervariable region when used herein refers to the amino acid residues of an antibody that are responsible for antigen binding.
  • the hypervariable region generally comprises amino acid residues from a "complementarity-determining region” or "CDR" (residues 24-34 (L1), 50-56 (L2) and 89-97 (L3) in the light-chain variable domain and 31 -35 (H1 ), 50-65 (H2) and 95-102 (H3) in the heavy-chain variable domain; (Kabat et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No.
  • CDR complementarity-determining region
  • phrases such as "Kabat position”, “variable domain residue numbering as in Kabat” and “according to Kabat” herein refer to this numbering system for heavy chain variable domains or light chain variable domains.
  • the actual linear amino acid sequence of a peptide may contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a FR or CDR of the variable domain.
  • a heavy chain variable domain may include a single amino acid insert (residue 52a according to Kabat) after residue 52 of CDR H2 and inserted residues (e.g. residues 82a, 82b, and 82c, etc. according to Ka- bat) after heavy chain FR residue 82.
  • the Kabat numbering of residues may be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a "standard” Kabat numbered sequence.
  • Framework region or "FR” residues are those VH or VL residues other than the CDRs as herein defined.
  • an “epitope” or “binding site” is an area or region on an antigen to which an antigen- binding peptide (such as an antibody) specifically binds.
  • a protein epitope may comprise amino acid residues directly involved in the binding (also called the immunodominant component of the epitope) and other amino acid residues, which are not directly involved in the binding, such as amino acid residues which are effectively blocked by the specifically antigen binding peptide (in other words, the amino acid residue is within the "solvent-excluded surface” and/or "footprint” of the specifically antigen binding peptide).
  • epitope herein includes both types of amino acid binding sites in any particular region of a hNKG2D that specifically binds to an anti-hNKG2D antibody, or another hNKG2D-specific agent according to the invention, unless otherwise stated (e.g., in some contexts the invention relates to antibodies that bind directly to particular amino acid residues).
  • NKG2Ds may comprise a number of different epitopes, which may include, without limitation, (1) linear peptide antigenic determinants, (2) conformational antigenic determinants which consist of one or more noncontiguous amino acids located near each other in a mature NKG2D conformation; and (3) post-translational antigenic determinants which consist, either in whole or part, of molecular structures covalently attached to a NKG2D, such as carbohydrate groups.
  • conformational antigenic determinants comprise NKG2D amino acid residues within about 4 A distance from an atom of an antigen-binding peptide.
  • the "solvent excluded surface” is the area of a molecule which, in a computer calcu- lation, cannot be reached by any water molecule, e.g., because of binding of the molecule to a ligand (Lee and Richards, J MoI Biol 1971 ;55:379-400, which is incorporated herein by reference).
  • an antibody of interest e.g., MS or 21 F2
  • an antibody of interest e.g., MS or 21 F2
  • a “paratope” is an area or region of an antigen-binding portion of an antibody that specifically binds an antigen. Unless otherwise stated or clearly contradicted by context, a paratope may comprise amino acid residues directly involved in epitope binding, several of which are typically in CDRs, and other amino acid residues, which are not directly involved in the binding, such as amino acid residues which are effectively blocked by the specifically bound antigen (in other words, the amino acid residue is within the "solvent-excluded surface” and/or "footprint” of the specifically bound antigen).
  • an anti-NKG2D antibody to "block" the binding of a NKG2D molecule to a natural NKG2D-ligand (e.g., MICA), means that the antibody, in an assay using soluble or cell-surface associated NKG2D and ligand molecules, can detectably reduce the binding of a NKG2D-molecule to the ligand in a dose-dependent fashion, where the NKG2D molecule detectably binds to the ligand in the absence of the antibody.
  • An exemplary assay for determining whether an anti-NKG2D antibody is capable of blocking MICA-binding is provided in Example 3. The same assay can be used for testing antibody-mediated blocking of other NKG2D ligands.
  • a “variant" of a polypeptide refers to a polypeptide having an amino acid sequence that is substantially identical to a reference polypeptide, typically a native or “parent” polypeptide.
  • the polypeptide variant may possess one or more amino acid substitutions, deletions, and/or insertions at certain positions within the native amino acid sequence and/or additions at one or both termini.
  • substantially identical in the context of two amino acid sequences means that the sequences, when optimally aligned, such as by the programs GAP or BEST- FIT using default gap weights, share at least about 50 percent sequence identity. Typically sequences that are substantially identical will exhibit at least about 60, at least about 70, at least about 80, at least about 90, at least about 95, at least about 98, or at least about 99 percent sequence identity.
  • a nucleic acid sequence (or element) is "operably linked" to another nucleic acid se- quence (or element) when it is placed into a functional relationship with the other nucleic acid sequence.
  • DNA for a pre-sequence or secretory leader is operably linked to DNA for (i.e. , coding for expression of) a polypeptide if it is expressed as a pre-protein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome-binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • operably linked means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, some elements, such as enhancers, do not have to be contiguous with a coding sequence in order to be operably linked. Linking typically is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers may be used in accordance with conventional practice.
  • an "isolated" molecule is a molecule that is the predominant species in the composition wherein it is found with respect to the class of molecules to which it belongs (i.e. , it makes up at least about 50% of the type of molecule in the composition and typically will make up at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or more of the species of molecule, e.g., peptide, in the composition).
  • a composition of an antibody molecule will exhibit 98%, 98%, or 99% homogeneity for antibody molecules in the context of all present peptide species in the composition or at least with respect to substantially active peptide species in the context of proposed use.
  • treatment refers to preventing, alleviating, managing, curing or reducing one or more symptoms or clinically relevant manifestations of a disease or disorder, unless contradicted by context.
  • treatment of a patient in whom no symptoms or clinically relevant manifestations of a disease or disor- der have been identified is preventive or prophylactic therapy, whereas clinical, curative, or palliative "treatment” of a patient in whom symptoms or clinically relevant manifestations of a disease or disorder have been identified generally does not constitute preventive or prophylactic therapy.
  • Each form of treatment may be considered a distinct aspect of the invention.
  • the present invention is based, in part, on anti-NKG2D antibodies with properties suitable for treating human patients suffering from NKG2D-related conditions, such as, e.g., autoimmune and inflammatory diseases and disorders.
  • Antibodies of the invention are typically either fully human or humanized in order to minimize the risk for an immune response against the antibody by the patient's own immune system, and bind to hNKG2D in its active form, i.e., a homodimer on the surface of a cell and associated with DAP10.
  • the antibodies of the invention are typically useful for treatment of conditions where NKG2D activity should be reduced.
  • Such antibodies can reduce or inhibit activation of NKG2D-expressing NK and/or T cells by, e.g., competing with or blocking one or more endogenous NKG2D-ligands for binding to NKG2D, down-modulating or otherwise reducing the amount of cell-surface NKG2D upon binding, and/or eliciting an ADCC or CDC response against the cells.
  • antibodies of the invention are antagonists and compete with one or more natural ligands such as MICA for binding to human NKG2D, thereby reducing ligand- induced NKG2D-activation.
  • MICA molecules have been clearly implicated in inflammatory diseases, and, as shown in Example 3, several human antibodies were effective at blocking MICA-binding to cell-surface NKG2D, particularly MS and 21 F2, and epitope determination showed that MS Fab obstructed MICA from binding (Example 11 , Figure 20). Both MS and 21 F2 were also highly efficient in blocking NK-cell mediated cytotoxicity (Example 6). Thus, these results demonstrate that the invention provides antibodies having such properties.
  • antibodies of the invention are efficient antagonists, but also have insignificant agonistic effect on hNKG2D signalling, thus not contributing to NKG2D-driven inflammation.
  • hNKG2D signalling no co-stimulation of immobilized MS on CD3-triggered proliferation of PBMCs could be detected, whereas immobi- lized ON72 resulted in a small but significant co-stimulation.
  • this difference may at least in part be due to the differences in epitopes, shown in Figures 20- 22.
  • bivalent MS antibody binds strongly to one monomer in an hNKG2D dimer complex, but blocks binding of a second MS antibody (or a second antigen- binding portion of the same antibody) to the second monomer.
  • an antigen- binding portion of a bivalent hzON72 antibody binds a first monomer in an hNKG2D dimer, it does not block the binding of a second hzON72 antibody (or a second antigen-binding portion of the same antibody) to the second monomer.
  • the invention provides human or humanized anti-NKG2D antibodies which, when added to NKG2D-expressing NK or T cells, cross-link not more than 2 hNKG2D dimers.
  • such antibodies are bivalent.
  • a bivalent antibody (such as, e.g., MS) for which the binding of the antigen-binding portion to an NKG2D monomer unit blocks further binding to the second NKG2D monomer unit can at most crosslink 2 hNKG2D dimers only.
  • a bivalent antibody which can bind an NKG2D monomer unit in an hNKG2D dimer without blocking binding to the second NKG2D monomer unit in an hNKG2D dimer can result in cross-linking of any number of hNKG2D dimers. Clustering of surface receptors commonly occurs in receptor activation.
  • the invention provides human or humanized anti-NKG2D antibodies which, when added to NKG2D-expressing NK or T cells, binds strongly only to one monomer in an hNKG2D dimer complex. Without being limited to theory, strong binding to both monomers of the dimer can be a prerequisite for activation of the NKG2D-receptor.
  • the ratio of the solvent-excluded surface areas from the first and second NKG2D monomer units by the binding of an antibody of the invention is more than about 1 :1 , at least about 2:1 , or at least about 3: 1.
  • the invention provides human or humanized anti-NKG2D antibodies which bind essentially the same epitope as MS. Without being limited to theory, inter- actions of a ligand with particular residues, or residue combinations, on the hNKG2D dimer could avoid or minimize agonist activity.
  • the epitope of an antibody of the invention comprises at least one residue selected from, at least 3 residues selected from, at least 5 residues selected from, at least 8 residues selected from, at least 10 residues selected from, at least 12 residues selected from, or all of the residues se- lected from the group consisting of Lys 150, Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, GIn 185, Leu 191 , Lys 197, Tyr 199, GIu 201 , Thr 205, Pro 206, Asn 207 and Thr 208 of hNKG2D (SEQ ID NO: 2).
  • the present invention provides a fully human antibody, or antigen- binding fragment thereof, that effectively prevents NKG2D-mediated cytotoxicity of a hNKG2D-expressing NK or T cell, competes with at least MICA in binding to hNKG2D; reduces the amount of cell-surface hNKG2D upon binding via, e.g. , stimulating down- modulation of hNKG2D, internalization of hNKG2D and/or preventing reappearance of hNKG2D; has an affinity to hNKG2D of 10 nM or less, cross-reacts with cynomolgus and/or rhesus NKG2D; and is non-depleting, e.g.
  • the antibody is a non-depleting fully human antibody of the lgG4 isotype, with an affinity to hNKG2D of 1 nM or less, preferably 300 pM or less, which blocks at least 50%, at least 70%, or at least 90% of endogenous hNKG2D-ligand binding, and reduces the amount of cell-surface hNKG2D with at least 10%, at least 30%, or at least 50%.
  • the antibody is a bivalent non-depleting fully human antibody of the lgG4 isotype, with an affinity below 100 pM, which has an EC50 concentration below 0.01 ng/ml for blocking the binding of full saturation dose of MICA-Fc to cell-surface associated NKG2D, is capable of reducing the amount of cell-surface NKG2D with at least 75% upon binding, and, optionally, has an EC50 concentration for reducing a ligand-induced NK cell cytotoxicity that is lower than the EC50 concentration required for binding to cell-curface associated NKG2D.
  • the antibody may further be capable of achieving, in an assay using NKG2D- expressing cells, its maximum level of hNKG2D down-modulation at a concentration lower than that required to obtain saturation of the hNKG2D receptors (i.e. , saturation dose).
  • the antibodies of the invention are characterized by particular functional and/or structural features or properties. Assays to evaluate the functional activities of anti-hNKG2D antibodies are described in detail in the Examples, and structural properties such as, e.g., amino acid sequences, are described below. Functional properties
  • the antibodies of the invention bind to hNKG2D.
  • an antibody of the invention binds to hNKG2D with high affinity, for example with a KD of 10 '7 M or less, a KD of 10 ⁇ 8 M or less, a KD of 1 nM or less, a KD of 0.3 nM or less, a KD of 0.2 nM or less, 0.1 nM or less, 0.05 nM or less, or 0.01 nM or less.
  • the antibody binds to hNKG2D with an affinity of 0.1 nM or less.
  • the invention provides antibodies also binding to one or more NKG2D orthologs in monkey such as cynomolgous monkey (Macaca fascicularis, NCBI accession No. AJ426429) and rhesus monkey (Macaca mulatta, NCBI accession No. AJ554302), and/or to hNKG2D homodimer, correctly folded monomeric full-length hNKG2D, hNKG2D fragment comprising an extracellular portion of hNKG2D, denatured hNKG2D, or to any combination of the preceding NKG2D forms.
  • monkey such as cynomolgous monkey (Macaca fascicularis, NCBI accession No. AJ426429) and rhesus monkey (Macaca mulatta, NCBI accession No. AJ554302)
  • hNKG2D homodimer homodimer
  • hNKG2D homodimer correctly folded monomeric full-length hNKG2D,
  • an antibody of the invention binds to cynomolgous and/or rhesus NKG2D with similar affinity or efficacy as it binds to hNKG2D.
  • an antibody can bind to NKG2D-expressing cynomolgous or rhesus NK or T cells with an EC50 of about 50% or more, about 65% or more, or about 75% or more, of the corresponding EC50 for a corre- sponding population of NKG2D-expressing human NK or T cells.
  • an antibody can bind to cynomolgous or rhesus NKG2D with an affinity of about 30% or more, about 50% or more, about 65% or more, or about 75% or more, about 80% or more, about 85% or more, or about 90% or more, of the affinity for hNKG2D.
  • Such antibodies have the advantage of allowing for toxicity testing in the most suitable animal model (or models) prior to use in humans.
  • antibodies of the invention also bind a form of NKG2D that known murine anti-hNKG2D antibodies such as ON72 do not bind. Specifically, as described in Example 3, pre-incubation with ON72 only blocked about 82% of subsequently added human 16F16 antibody from binding to hNKG2D, while pre-incubation with 16F16 blocked about 95% of subsequently added ON72 from binding to hNKG2D. Furthermore, the antibodies of the invention can reduce or inhibit hNKG2D-mediated activation of NK or T cells, i.e., antagonize the hNKG2D receptor. This may be tested in, e.g., one or more cytotoxicity assays described herein or known in the art.
  • an antibody inhibits hNKG2D-mediated activation of an NK or T cell if it inhibits the NK- or T cell- mediated killing of an NKG2D-ligand-expressing target cell by at least 10%, more preferably by at least 30%, even more preferably by at least 40%, at least 50%, at least 60%, at least 70%, at least 80% or at least 90%, as compared to target cell killing in the absence of any anti-hNKG2D antibody or in the presence of a non-specific, control antibody.
  • Antibodies of the invention that are hNKG2D antagonists can have no or low agonist activity.
  • such antibodies are human or humanized.
  • Agonist activity may be tested in one of the assay described herein, or an assay known in the art.
  • one type of assay is a co-stimulation assay measuring proliferation of peripheral blood lymphocytes (PBMCs) stimulated with low levels of CD3 in the presence or absence of immobilized anti- NKG2D antibody (see Example 10).
  • proliferation in the presence of an an- tibody of the invention is not more than 30%, not more than 20%, not more than 10%, not more than 5% or not significantly higher than in the absence of antibody.
  • proliferation in the presence of an antibody of the invention is not significantly higher than in the absence of antibody.
  • hNKG2D agonist activity of an antibody of the invention in an agonist assay is not more than 30%, not more than 20%, not more than 10%, not more than 5%, or not significantly higher than a control value.
  • the control is preferably a negative control, such as, e.g., in the absence of antibody, in the absence of cell or another reagent, and/or in the presence of an irrelevant antibody.
  • agonist activity of an antibody of the invention is not significantly higher than a control value.
  • the invention provides antibodies that have a lower, preferably substantially lower, EC50 concentration for blocking ligand-induced cytotoxicity than for binding to cell-surface NKG2D of an NK or T cell.
  • the EC50 concentration for binding to cell-surface NKG2D expressed on BaF/3 cells was similar to the EC50 concentration for blocking NK-cell mediated killing of ligand- (ULBP3-) expressing target cells (0.065 ⁇ g/ml), whereas 21 F2 had a lower, and MS a substantially lower, EC50 for blocking cytotoxicity (21 F2: 0.021 ⁇ g/ml; MS: 0.012 ⁇ g/ml) than for binding to cell-surface NKG2D (21 F2: 0.033 ⁇ g/ml; MS: 0.032 g/ml) (see Examples 6 and 9).
  • the invention provides antibodies, preferably human or humanized antibodies, that have a lower EC50 concentration for blocking ligand-induced cytotoxicity than for binding to cell-surface NKG2D of an NK or T cell.
  • the EC50 for blocking cytotoxicity of NK or T cells of a cell line or other suitable preparation can be, e.g., about 95% or less, about 90% or less, about 85% or less, about 80% or less, about 70% or less, about 50% or less, or about 40% or less, of the EC50 for binding to cell-surface NKG2D of the same cell line or preparation.
  • Exemplary cell lines for testing include NK-92 and NKL cells.
  • the invention provides antibodies that achieve maximum blockage of NK cell cytotoxicity at a concentration lower than the concentration required to saturate the available hNKG2D-receptors.
  • the antibodies also compete with MS in binding to hNKG2D.
  • such antibodies bind to essentially the same hNKG2D epitope as MS.
  • the antibodies may reduce or inhibit NKG2D-mediated activation by, e.g., interfering with the hNKG2D-binding of one or more endogeous hNKG2D-ligands.
  • the antibodies may reduce or inhibit the hNKG2D-binding of MICA; MICB; ULBP1 ; ULBP2; ULBP4; and/or RAET1-family member; e.g., by reducing or inhibiting the hNKG2D-binding of MICA; or of MICA and MICB; or of MICA and ULBP3; or of MICA, MICB, and ULBP3; or of MICA, MICB, and all ULBP1 , -2, -3, and 4; or of MICA, MICB, and one or more RAET1 family members.
  • antibodies of the invention are capable of inhibiting at least 30% of ligand binding, or at least 50% of ligand binding, or at least 70% of ligand binding, or at least 80%, or at least 90% of ligand binding.
  • the IC50 for an antibody of the invention to inhibit the hNKG2D-binding of 1 ⁇ g MICA-mFc is 1 nM or less, 0.5 nM or less, 0.2 nM or less, 0.1 nM or less, 0.05 nM or less, or 0.02 nM or less, 0.01 nM or less, 0.005 or less, or 0.002 or less.
  • full blockage of 1 ⁇ g MICA-mFc binding is achieved at an antibody concentration of 5 nM or less, 1 nM or less, 0.7 nM or less, 0.5 nM or less, or 0.2 nM or less, 0.1 nM or less, 0.05 nM or less, or about 0.02 nM or less.
  • the invention provides antibodies, especially human antibodies, that are as efficient or more efficient in reducing or inhibiting ligand hNKG2D-binding, such as, e.g., MICA binding to hNKG2D, than any of ON72, BAT221 , 5C6, 1 D11 , ECM217, and 149810.
  • an anti-hNKG2D antibody of the invention can be capable of reducing the amount of cell-surface hNKG2D upon (i.e., following) binding.
  • Reduction of cell-surface associated hNKG2D upon binding of an antibody can be an advantageous feature, since it reduces the number of hNKG2D receptors available for ligand binding and subsequent activation. Without being limited to theory, this reduction may be caused by NKG2D down-modulation, internalization, or other mechanism.
  • anti- hNKG2D antibodies having a human Fc-region such as human antibodies, are capable of effectively reducing the amount of cell-curface hNKG2D.
  • human anti-hNKG2D antibodies 16F16, MS, and 21 F2 all reduced the amount of cell-surface hNKG2D with about 75% or more after overnight incubation in the absence of serum, with MS being the most effective, achieving 75-90% downmodulation at a low concentration ( Figures 15-17).
  • an MS concentration corresponding to less than saturating concentration on hNKG2D-expressing BaF/3 cells achieved maximum downmodulation ( Figure 16B).
  • the invention provides antibodies binding to hNKG2D that are able to achieve maximum down-modulation of hNKG2D at less than saturating concentrations.
  • such antibodies also compete with MS in binding to hNKG2D.
  • such antibodies also bind to essentially the same hNKG2D epitope as MS.
  • An antibody of the invention can be capable of reducing cell surface hNKG2D by at least 10%, at least 20%, at least 30%, at least 50%, at least 70%, or at least 90% as compared to cell-surface hNKG2D in the absence of anti-hNKG2D antibody or in the presence of a non-specific control antibody.
  • the antibodies achieve reduction of cell-surface NKG2D while causing no or minimal activation of NKG2D-receptor signalling, i.e. , with no or minimal agonist activity.
  • an anti-hNKG2D antibody of the invention can be capable of achieving maximum down-modulation of cell-surface NKG2D expressed by a cell or cell-line at a concentration lower than a saturating concentra- tion.
  • the invention provides antibodies that compete with and/or bind to the same epitope on hNKG2D as 16F16, 16F31 , MS, and/or 21 F2, more preferably MS and/or 21 F2.
  • Such antibodies can be identified based on their ability to cross-compete with 16F16, 16F31 , MS, or 21 F2 in standard hNKG2D binding assays as described herein.
  • test antibody to inhibit the binding of 16F16, 16F31 , MS, or 21 F2 to hNKG2D demonstrates that the test antibody can compete with 16F16, 16F31 , MS, or 21 F2 for binding to hNKG2D and thus can bind to the same epitope on hNKG2D as 16F16, 16F31 , MS, or 21 F2.
  • the antibody that binds to the same epitope on hNKG2D as 16F16, 16F31 , MS or 21 F2 is a human monoclonal antibody. Such human monoclonal antibodies can be prepared and isolated as described in the Examples.
  • the antibody binds to a different epitope than any of the mouse monoclonal antibodies ON72, BAT221 , 5C6, 1 D1 1 , ECM217, and 149810, and cross-competes more with 16F16, 16F31 , MS, or 21 F2 than with either of the listed mouse monoclonal antibodies.
  • the epitope of an antibody of the invention comprises one or more residues selected from Lys 150, Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, GIn 185, Leu 191 , Lys 197, Tyr 199, GIu 201 , Thr 205, Pro 206, Asn 207 and Thr 208 of hNKG2D (SEQ ID NO: 2).
  • the epitope of an antibody of the invention comprises 5 or more residues selected from Lys 150, Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, GIn 185, Leu 191 , Lys 197, Tyr 199, GIu 201 , Thr 205, Pro 206, Asn 207 and Thr 208 of hNKG2D (SEQ ID NO: 2).
  • the epitope of an antibody of the invention comprises 8, 10, 12 or more residues selected from Lys 150, Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, GIn 185, Leu 191 , Lys 197, Tyr 199, GIu 201 , Thr 205, Pro 206, Asn 207 and Thr 208 of hNKG2D (SEQ ID NO: 2).
  • the epitope of an antibody of the invention comprises the residues Lys 150, Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, GIn 185, Leu 191 , Lys 197, Tyr 199, GIu 201 , Thr 205, Pro 206, Asn 207 and Thr 208 of hNKG2D (SEQ ID NO: 2).
  • the epitope of an antibody of the invention consists essentially of the residues Lys 150, Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, GIn 185, Leu 191 , Lys 197, Tyr 199, GIu 201 , Thr 205, Pro 206, Asn 207 and Thr 208 of hNKG2D (SEQ ID NO: 2).
  • the epitope of an antibody of the invention consists of one or more residues selected from Lys 150, Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, GIn 185, Leu 191 , Lys 197, Tyr 199, GIu 201 , Thr 205, Pro 206, Asn 207 and Thr 208 of hNKG2D (SEQ ID NO: 2).
  • the epitope of an antibody of the invention consists of the residues Lys 150, Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met
  • the epitope of an antibody of the invention comprises one or more residues involved in hydrogen-binding selected from Lys 150, Ser 151 , Tyr 152, Ne 181 , Met 184, GIn 185, Lys 197, Thr 205, and Asn 207 of hNKG2D (SEQ ID NO: 2). In one embodiment, the epitope of an antibody of the invention comprises 5 or more residues involved in hydrogen-binding selected from Lys 150, Ser 151 , Tyr 152, Ne 181 , Met 184, GIn
  • the epitope of an antibody of the invention comprises Lys 150, Ser 151 , Tyr 152, Ne 181 , Met 184, GIn 185, Lys 197, Thr 205, and Asn 207 of hNKG2D (SEQ ID NO: 2).
  • Preferred antibodies of the invention exhibit at least one, more preferably two, three, four, five or more, of the following properties: (a) prevents NKG2D-mediated activation of an NKG2D-expressing NK or T cell, optionally with an EC50 for reducing ligand-induced cytotoxicity lower than the EC50 for binding to the cell; (b) competes with at least one NKG2D ligand in binding to NKG2D, preferably with at least MICA and ULBP3; (c) reduces the amount of NKG2D on the surface of a NKG2D-expressing NK or T cell, preferably with at least 75%; (d) binds to cynomolgous and/or rhesus NKG2D, preferably with no less than 50% of the affinity by which it binds to hNKG2D; (e) binds to more than one form or conformation of NKG2D; (f) binds to NKG2D with a Kd of 1 nM
  • Preferred antibodies of the invention are the human monoclonal antibodies 16F16,
  • Certain anti-NKG2D antibodies of the invention has the same or a similar paratope as MS.
  • the antibody has a paratope comprising residues corresponding to one or more of Tyr 33 and Trp 97 of the MS L chain (SEQ ID NO: 41), and/or to one or more of GIn 1 , Asp 26, Asp 27, Ser 30, Ser 31 , Tyr 32, Tyr 33, His 50, Ser 52, Tyr 53, Ser 54, Ser 56, Ala 57, Asn 58, Trp 98 and Asp 99 of the MS H chain (SEQ ID NO: 40).
  • the antibody has a paratope comprising residues corresponding to Tyr 33 and Trp 97 of the MS L chain (SEQ ID NO: 41), and/or to 3, 5, 7, 10 or more of GIn 1 , Asp 26, Asp 27, Ser 30, Ser 31 , Tyr 32, Tyr 33, His 50, Ser 52, Tyr 53, Ser 54, Ser 56, Ala 57, Asn 58, Trp 98 and Asp 99 of the MS H chain (SEQ ID NO: 40).
  • the antibody has a paratope comprising residues corresponding to Tyr 33 and Trp 97 of the MS L chain (SEQ ID NO: 41), and GIn 1 , Asp 26, Asp 27, Ser 30, Ser 31 , Tyr 32, Tyr 33, His 50, Ser 52, Tyr 53, Ser 54, Ser 56, Ala 57, Asn 58, Trp 98 and Asp 99 of the MS H chain (SEQ ID NO: 40).
  • the antibody has a paratope consisting essentially of resi- dues corresponding to Tyr 33 and Trp 97 of the MS L chain (SEQ ID NO: 41 ), and GIn 1 , Asp 26, Asp 27, Ser 30, Ser 31 , Tyr 32, Tyr 33, His 50, Ser 52, Tyr 53, Ser 54, Ser 56, Ala 57, Asn 58, Trp 98 and Asp 99 of the MS H chain (SEQ ID NO: 40).
  • the antibody has a paratope consisting of residues corresponding to Tyr 33 and Trp 97 of the MS L chain (SEQ ID NO: 41), and GIn 1 , Asp 26, Asp 27, Ser 30, Ser 31 , Tyr 32, Tyr 33, His 50, Ser 52, Tyr 53, Ser 54, Ser 56, Ala 57, Asn 58, Trp 98 and Asp 99 of the MS H chain (SEQ ID NO: 40).
  • SEQ ID NO: 41 the amino acid chain
  • GIn 1 Asp 26, Asp 27, Ser 30, Ser 31 , Tyr 32, Tyr 33, His 50, Ser 52, Tyr 53, Ser 54, Ser 56, Ala 57, Asn 58, Trp 98 and Asp 99 of the MS H chain (SEQ ID NO: 40).
  • the hNKG2D-binding of such "mixed and matched" antibodies can be tested using the binding assays described herein (e.g. flow cytometry, Biacore, ELISAs) and/or using a cytotoxicity assay as described herein.
  • a VH sequence from a particular VH/VL pairing is replaced with a structurally similar VH sequence.
  • a VL sequence from a particular VH/VL pairing is replaced with a structurally similar VL sequence.
  • the invention provides an isolated monoclonal antibody, or antigen binding portion thereof, comprising: (a) a VH region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 11 , 13, 44, and 46, and (b) a VL region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 12, 14, 45, and 47; wherein the antibody binds hNKG2D.
  • Preferred heavy and light chain combinations include: (a) a VH region comprising the amino acid sequence of SEQ ID NO: 11 ; and (b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 12; (a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 13; and (b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 14; (a) a VH region comprising the amino acid sequence of SEQ ID NO: 44; and (b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 46; or (a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 45; and (b) a light chain variable region comprising the amino acid sequence of SEQ ID NO: 47.
  • the invention provides antibodies that comprise the heavy chain and light chain CDRIs, CDR2s and/or CDR3s of 16F16, 16F31 , MS, or 21 F2, or combinations thereof.
  • the CDR regions are delineated using the Kabat system (Kabat et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). See, e.g., Figures 4 and 5.
  • VH CDR1 , 2 and 3 sequences and VL CDR1 , 2 and 3 sequences can be "mixed and matched" (i.e., CDRs from different antibodies can be mixed and match, although each antibody can contain a VH CDR1 , 2 and 3 and a VL CDR1 , 2 and 3) to create other anti-hNKG2D binding molecules of the invention.
  • the hNKG2D- binding of such "mixed and matched" antibodies can be tested using the binding assays described above and in the Examples (e.g.
  • the CDR1 , CDR2 and/or CDR3 se- quence from a particular VH sequence is replaced with a structurally similar CDR se- quence(s).
  • the CDR1 , CDR2 and/or CDR3 sequence from a particular VL sequence preferably is replaced with a structurally similar CDR sequence(s).
  • the VL CDRIs and CDR3s of 16F16, 16F31 , MS, and 21 F2 and the VL CDR2 sequences of MS and 21 F2 share some structural similarity and therefore are amenable to mixing and matching.
  • VH and VL sequences can be created by substituting one or more VH and/or VL CDR region sequences with structurally similar sequences from the CDR sequences disclosed herein for monoclonal antibodies antibodies 16F16, 16F31 , MS, and 21 F2.
  • the invention provides an isolated monoclonal antibody, or antigen binding portion thereof comprising: (a) a VH CDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 15, 21 , 48, and 54; (b) a VH CDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 16 , 22, 49, and 55; (c) a VH CDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 17, 23, 50, and 56; (d) a VL CDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:18, 24, 51 , and 57; (e) a VL CDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:19, 25, 52, and 57; and (f) a VL CDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 20, 26, 53, and 59; where
  • the antibody comprises: (a) a VH CDR1 comprising SEQ ID NO:15; (b) a VH CDR2 comprising SEQ ID NO:16; (c) a VH CDR3 comprising SEQ ID NO: 17; (d) a VL CDR1 comprising SEQ ID NO:18; (e) a VL CDR2 comprising SEQ ID NO: 19; and (f) a VL CDR3 comprising SEQ ID NO: 20.
  • the antibody comprises: (a) a VH CDR1 comprising SEQ ID NO: 21 ; (b) a VH CDR2 comprising SEQ ID NO:22; (c) a VH CDR3 comprising SEQ ID NO:23; (d) a VL region CDR1 comprising SEQ ID NO:24; (e) a VL CDR2 comprising SEQ ID NO:25; and (f) a VL CDR3 comprising SEQ ID NO: 26.
  • the antibody comprises: (a) a VH CDR1 compris- ing SEQ ID NO: 48; (b) a VH CDR2 comprising SEQ ID NO:49; (c) a VH CDR3 comprising SEQ ID NO:50; (d) a VL region CDR1 comprising SEQ ID NO:51 ; (e) a VL CDR2 comprising SEQ ID NO:52; and (f) a VL CDR3 comprising SEQ ID NO: 53.
  • the antibody comprises: (a) a VH CDR1 comprising SEQ ID NO: 54; (b) a VH CDR2 comprising SEQ ID NO:55; (c) a VH CDR3 comprising SEQ ID NO:56; (d) a VL region CDR1 comprising SEQ ID NO:57; (e) a VL CDR2 comprising SEQ ID NO:58; and (f) a VL CDR3 comprising SEQ ID NO: 59.
  • the antibody comprises: (a) a VH CDR1 consisting of SEQ ID NO:15; (b) a VH CDR2 consisting of SEQ ID NO:16; (c) a VH CDR3 consisting of SEQ ID NO:17; (d) a VL CDR1 consisting of SEQ ID NO:18; (e) a VL CDR2 consisting of SEQ ID NO: 19; and (f) a VL CDR3 consisting of SEQ ID NO: 20.
  • the antibody comprises: (a) a VH CDR1 consisting of SEQ ID NO: 21 ; (b) a VH CDR2 consisting of SEQ ID NO:22; (c) a VH CDR3 consisting of SEQ ID NO:23; (d) a VL region CDR1 consisting of SEQ ID NO:24; (e) a VL CDR2 consisting of SEQ ID NO:25; and (f) a VL CDR3 consisting of SEQ ID NO: 26.
  • the antibody comprises: (a) a VH CDR1 consisting of SEQ ID NO: 48; (b) a VH CDR2 consisting of SEQ ID NO:49; (c) a VH CDR3 consisting of SEQ ID NO:50; (d) a VL region CDR1 consisting of SEQ ID NO:51 ; (e) a VL CDR2 consisting of SEQ ID NO:52; and (f) a VL CDR3 consisting of SEQ ID NO: 53.
  • the antibody comprises: (a) a VH CDR1 consisting of SEQ ID NO: 48; (b) a VH CDR2 consisting of SEQ ID NO:49; (c) a VH CDR3 consisting of SEQ ID NO:50; (d) a VL region CDR1 consisting of SEQ ID NO:51 ; (e) a VL CDR2 consisting of SEQ ID NO:52; and (f) a VL CDR3 consisting of SEQ ID NO: 53, and residues corresponding to one, two, or all of GIn 1 , Asp 26, and Asp 27 in the MS H chain (SEQ ID NO: 40).
  • the antibody comprises: (a) a VH CDR1 consisting of SEQ ID NO: 54; (b) a VH CDR2 consisting of SEQ ID NO:55; (c) a VH CDR3 consisting of SEQ ID NO:56; (d) a VL region CDR1 consisting of SEQ ID NO:57; (e) a VL CDR2 consisting of SEQ ID NO:58; and (f) a VL CDR3 consisting of SEQ ID NO: 59.
  • an antibody of the invention comprises a VH region from a particular germline H chain immunoglobulin gene, or a combination of particular germline H chain immunoglobulin genes; and/or a VL region from a particular germline L chain immunoglobulin gene, or a combination of particular germline L chain immunoglobulin genes.
  • Such combinations can be obtained, e.g., in vivo via somatic recombination in a B cell.
  • the invention provides an isolated anti-hNKG2D antibody, or an antigen-binding fragment thereof, wherein the antibody: (a) comprises a VH region from a human VH3_21 , VH3_20, VH4_59, or VH5_51 gene recombined with a human D3-9, D3-10, or D3_10_R3 gene and a JH3, JH4 or JH6 gene, (b) comprises a VL region derived from a human VKI_L15 or VKIII_A27 or VKIII_L6 gene recombined with a human JK1 , JK2 or JK3 gene, and (c) the antibody binds to hNKG2D.
  • the antibody comprises a VH region from a human VH3_21 , VH3_20, VH4_59, or VH5_51 gene recombined with a human D3-9, D3-10, or D3_10_R3 gene and a J
  • the invention provides an isolated anti-hNKG2D antibody, or an antigen-binding fragment thereof, comprising a VH region obtained by a recombination of human VH3_21 , D3-9, and JH4 genes and a VL region obtained by a recombination of human VKI_L15 and JK2 genes.
  • the invention provides an isolated anti-hNKG2D antibody, or an antigen-binding fragment thereof, comprising a VH region obtained by a recombination of human VH3_20, D3-10, and JH6 genes and a VL region obtained by a recombination of human VKIII_A27 and JK3 genes.
  • the invention provides an isolated anti-hNKG2D antibody, or an antigen-binding fragment thereof, comprising a VH region obtained by a recombination of human VH4_59, a D gene, and JH3 genes and a VL region obtained by a recombination of human VKIII_A27 and JK1 genes.
  • the invention provides an isolated anti-hNKG2D antibody, or an antigen-binding fragment thereof, comprising a VH region obtained by a recombination of human VH5_51 , D3_10_R3, and JH4 genes and a VL region obtained by a recombination of human VKIII_L6 and JK1 genes.
  • the invention provides isolated anti-NKG2D antbodies obtained by introducing one, two, three, four or more amino acid substitutions and/or somatic hypermutations in the VH and/or VL region of an anti-hNKG2D antibody de- scribed above.
  • a human antibody comprises heavy or light chain variable regions "of” or “derived from” or that are “the product of” a particular germline sequence if the variable regions of the antibody are obtained from a system (as described below) that uses human germline immunoglobulin genes.
  • systems include immunizing a transgenic mouse carrying human immunoglobulin genes with the antigen of interest or screening a human immunoglobulin gene library displayed on phage with the antigen of interest.
  • a human antibody that is "of” or “derived from” or “the product of” a human germline immunoglobulin sequence can be identified as such by comparing the amino acid sequence of the human antibody to the amino acid sequences of human germline immunoglobulins and se- lecting the human germline immunoglobulin sequence that is closest in sequence (i.e., greatest % identity) to the sequence of the human antibody.
  • a human antibody that is "of” or “derived from” or “the product of” a particular human germline immunoglobulin sequence may contain amino acid differences as compared to the germline sequence, due to, for example, naturally-occurring somatic mutations or intentional introduction of site-directed muta- tion(s) (which may be selected substitutions).
  • a human antibody is typically at least 90% identical in amino acid sequence to an amino acid sequence encoded by a recombined germline immunoglobulin sequence and can usually be identified as human when compared to the germline immunoglobulin amino acid sequences of other species (e.g., murine germline sequences).
  • a human antibody may be at least 95%, or even at least 96%, 97%, 98%, or 99% identical in amino acid sequence to the amino acid sequence encoded by the recombined germline immunoglobulin gene.
  • a human antibody derived from a particular human germline sequence will display no more than 10 amino acid differences from the amino acid sequence encoded by the human germline immunoglobulin gene.
  • the human antibody may display no more than 8, no more than 5, or even no more than 4, 3, 2, or 1 amino acid difference, or no amino acid difference, from the amino acid sequence encoded by the recombined germline immunoglobulin gene.
  • an antibody of the invention comprises heavy and light chain variable regions comprising amino acid sequences that are homologous to the amino acid sequences of the preferred antibodies described herein, and wherein the antibodies retain the desired functional properties of the anti-hNKG2D antibodies of the invention.
  • the invention provides an isolated antibody, or antigen binding portion thereof, comprising a heavy chain variable region and a light chain variable region, wherein: (a) the VH region comprises an amino acid sequence that is at least 80% identical to an amino acid sequence selected from the group consisting of SEQ ID NOs: 1 1 , 13, 44, and 46; (b) the VL region comprises an amino acid sequence that is at least 80% identical to an amino acid sequence selected from the group consisting of SEQ ID NOs: 12, 14, 45, and 47; (c) the antibody binds to hNKG2D and exhibits at least one of the functional properties described herein, preferably several of the functional properties described herein.
  • the VH and/or VL amino acid sequences may be 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the sequences set forth above.
  • An antibody having VH and VL regions having high (i.e. , 80% or greater) identity to the VH and VL regions of the sequences set forth above can be obtained by mutagenesis (e.g., site-directed or PCR- mediated mutagenesis) of nucleic acid molecules encoding SEQ ID NOs:11 -14 or 44-47, followed by testing of the encoded altered antibody for retained function (e.g., hNKG2D binding affinity, hNKG2D-ligand blocking, hNKG2D downmodulation, or reduction of NKG2D- mediated activation of an NK or T cell) using the functional assays described herein.
  • mutagenesis e.g., site-directed or PCR- mediated mutagenesis
  • the encoded altered antibody for retained function e.g.,
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm in sequence-analysis software. Protein analysis software matches similar sequences using measures of similarity assigned to various substitutions, deletions and other modifications, including conservative amino acid substitutions.
  • the percent identity between two amino acid sequences can be determined, e.g., using the Needleman and Wunsch (J. MoI. Biol. 48:444-453 (1970)) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1 , 2, 3, 4, 5, or 6.
  • Polypeptide sequences can also be compared using FASTA, applying default or recommended parameters.
  • FASTA e.g. , FASTA2 and FASTA3
  • FASTA3 provides alignments and percent sequence identity of the regions of the best over- lap between the query and search sequences (Pearson, Methods Enzymol. 1990; 183:63-98; Pearson, Methods MoI. Biol. 2000; 132: 185-219).
  • the percent identity between two amino acid sequences can also be determined using the algorithm of E. Meyers and W. Miller (Comput. Appl. Biosci., 1988;11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • Another algorithm for comparing a sequence to a other sequences contained in a database is the computer program BLAST, especially blastp, using default parameters. See, e.g., Altschul et al., J. MoI. Biol. 1990;215:403-410; Altschul et al., Nucleic Acids Res. 1997;25:3389-402 (1997); each herein incorporated by reference.
  • the protein sequences of the present invention can there be used as a "query sequence" to perform a search against public databases to, for example, identify related sequences. Such searches can be performed using the XBLAST program (version 2.0) of Altschul, et al. 1990 (supra).
  • Gapped BLAST can be utilized as described in Altschul et al., 1997 (supra).
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • XBLAST and NBLAST can be used. See http://www. ncbi.nlm.nih.gov.
  • an antibody of the invention comprises a VH region com- prising CDR1 , CDR2 and CDR3 sequences and a VL region comprising CDR1 , CDR2 and CDR3 sequences, wherein one or more of these CDR sequences comprise specified amino acid sequences based on the preferred antibodies described herein; 16F16, 16F31 , MS, or 21 F2, wherein one or more CDRs optionally contains one or more conservative amino acid modifications, and wherein the antibodies retain the desired functional properties of the anti- hNKG2D antibodies of the invention.
  • the invention provides an isolated antibody, or antigen-binding fragment thereof, comprising a heavy chain variable region comprising CDR1 , CDR2, and CDR3 sequences and a light chain variable region comprising CDR1 , CDR2, and CDR3 sequences, wherein: (a) the VH region CDR3 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 17, 23, 50, and 56; (b) the VL region CDR3 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 20, 26, 53 and 59; (c) one or more CDRs optionally contains one or more conservative amino acid modifications, and (d) the antibody binds to hNKG2D and exhibits at least one of the functional properties described herein, more preferably several of the functional properties de- scribed herein.
  • the VH region CDR2 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 16, 22, 49, and 55; and the VL region CDR2 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 19, 25, 52, and 58, wherein one or more CDRs optionally contains one or more conservative amino acid modifications.
  • the VH region CDR1 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 15, 21 , 48, and 54, and conservative modifications thereof; and the VL region CDR1 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs: 18, 24, 51 , and 57, wherein one or more CDRs optionally contains one or more conservative amino acid modifications.
  • conservative amino acid modifications is intended to refer to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody containing the amino acid sequence. Such conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into an antibody of the invention by standard techniques known in the art, such as, e.g., site-directed mutagenesis and PCR-mediated mutagenesis.
  • An antibody sequence comprising amino acid modifications as compared to a parent antibody is typically at least 90%, preferably at least 95%, 98%, or 99% identical to the corresponding amino acid sequence in the parent and/or comprises at most 10, preferably at most 5, 4, 3, 2 amino acid modifications as compared to the parent antibody sequence.
  • Constant amino acid substitutions are typically those in which an amino acid residue is replaced with an amino acid residue having a side chain with similar physico- chemical properties. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, ar- ginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g.
  • glycine asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, me- thionine
  • beta-branched side chains e.g. threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine.
  • amino acid residues within the CDR regions of an antibody of the invention can be replaced with other amino acid residues from the same side chain family and the altered antibody can be tested for retained function (i.e., the functions set forth in (c), (d) and (e) above) using the functional assays described herein.
  • Antigen-binding fragments i.e., the functions set forth in (c), (d) and (e) above.
  • anti-hNKG2D antibodies of the invention may be prepared as full-length antibodies or antigen-binding fragments thereof.
  • antigen-binding fragments include Fab, Fab', F(ab)2, F(ab')2, F(ab)3, Fv (typically the VL and VH domains of a single arm of an antibody), single-chain Fv (scFv; see e.g., Bird et al., Science 1988;242:423-426; and Huston et al.
  • dsFv, Fd typically the VH and CH 1 domain
  • dAb typically a VH domain
  • VH, VL, VhH, and V-NAR domains monovalent molecules comprising a single VH and a single VL chain
  • minibodies, diabodies, triabodies, tetrabodies, and kappa bodies see, e.g., Ill et al., Protein Eng 1997; 10:949-57
  • Antibody fragments can be obtained using conventional recombinant or protein engineering techniques, and the fragments can be screened for antigen-binding or other function in the same manner as are intact antibodies.
  • F(ab')2 fragments can be isolated directly from recombinant host cell culture.
  • the antibody of choice is a single-chain Fv fragment (scFv). See WO 1993/16185; U.S. Pat. No. 5,571 ,894; and U.S. Pat. No. 5,587,458.
  • the antibody fragment may also be a "linear antibody", e.g. , as described in U.S. Pat. No. 5,641 ,870, for example. Such linear antibody fragments may be monospecific or bispecific. Multispecific Molecules
  • the present invention features multispecific molecules comprising an anti-hNKG2D antibody, or an antigen-fragment thereof, of the invention.
  • Such multispecific molecules include bispecific molecules comprising at least one first binding specificity for hNKG2D and a second binding specificity for a second target epitope.
  • bispecific molecules are bispecific antibodies.
  • Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes. Methods for making bispecific antibodies are known in the art, and traditional production of full-length bispecific antibodies is usually based on the coexpression of two immunoglobulin heavy-chain-light- chain pairs, where the two chains have different specificities (Millstein et a/. , Nature, 305: 537-539 (1983)).
  • Bispecific antibodies can be prepared as full-length antibodies or antibody fragments (e.g. F(ab')2 bispecific antibodies) or any other antigen-binding fragments described herein.
  • the bispecific antibodies In the bispecific antibodies according to the present invention, at least one binding epitope is on the hNKG2D protein.
  • the anti-NKG2D-binding moiety may be combined with second moiety that binds to a molecule on a pro-inflammatory leukocyte, e.g., a T-cell receptor molecule (e.g. CD2, CD3, CD4, or CD8), so as to focus cellular defense mechanisms to a pro-inflammatory hNKG2D-expressing cell.
  • the bispecific antibodies can, e.g., be used to direct cytotoxic agents to, or an ADCC/CDC attack on, pro-inflammatory cells that express NKG2D.
  • the cytotoxic agent could be, e.g., saporin, an anti-interferon- alpha agent, a vinca alkaloid, the ricin A chain, methotrexate, or a radioactive isotope.
  • the second moiety binds a cell-associated target that is presented on or expressed by cells associated with a disease state normally regulated by effector lymphocytes, such as cancer, viral infection, or the like.
  • a typical target may be a cell stress-associated molecule such as a MIC molecule (e.g., MIC-A or MIC-B) or a ULBP (e.g., Rae-1 , H-60, ULBP2, ULBP3, HCMV UL18, or Rae-1 ⁇ ) or a pathogen- associated molecule such as a viral hemagglutinin.
  • multispecific molecules include those produced from the fusion of a hNKG2D- binding antibody moiety to one or more other non-antibody proteins.
  • Such multispecific pro- teins and how to construct them have been described in the art. See, e.g., Dreier et al. (Bio- conjug. Chem. 9(4): 482-489 (1998)); U.S. Patent 6,046,310; U.S. Patent Publication No. 20030103984; European Patent Application 1 413 316; US Patent Publication No. 20040038339; von Strandmann et al., Blood (2006; 107: 1955-1962.), and WO 2004056873.
  • the non-antibody protein could be, for example, a suitable ligand for any of the antigens of "second moiety" described I the preceding section; e.g., a ligand for a T-cell or Fc receptor, or a cell-stress molecule such as MIC-A, MIC-B, ULBP, or a pathogen-associated molecule such as a viral hemagglutinin.
  • a suitable ligand for any of the antigens of "second moiety" described I the preceding section e.g., a ligand for a T-cell or Fc receptor, or a cell-stress molecule such as MIC-A, MIC-B, ULBP, or a pathogen-associated molecule such as a viral hemagglutinin.
  • Multispecific molecules with more than two valencies are also contemplated.
  • trispecific antibodies can be prepared. Tutt et al., J. Immunol, 147: 60 (1991).
  • the multispecific molecules of the present invention can be prepared by conjugating the constituent binding specificities using methods known in the art. For example, each binding specificity of the multispecific molecule can be generated separately and then conjugated to one another. When the binding specificities are proteins or peptides, a variety of coupling or cross-linking agents can be used for covalent conjugation.
  • cross-linking agents examples include protein A, carbodiimide, N-succinimidyl-S-acetyl-thioacetate (SATA), 5,5'- dithiobis(2-nitrobenzoic acid) (DTNB), o-phenylenedimaleimide (oPDM), N-succinimidyl-3-(2- pyridyldithio)propionate (SPDP), and sulfosuccinimidyl 4-(N-maleimidomethyl) cyclohaxane- 1-carboxylate (sulfo-SMCC) (see e.g., Karpovsky et al. (1984) J. Exp. Med.
  • conjugating agents are SATA and sulfo-SMCC, both available from Pierce Chemical Co. (Rockford, IL).
  • binding specificities are antibodies, they can be conjugated via sulthydryl bonding of the C-terminus hinge regions of the two heavy chains.
  • the hinge region is modified to contain an odd number of sulfhydryl residues, preferably one, prior to conjugation.
  • both binding specificities can be encoded in the same vector and expressed and assembled in the same host cell.
  • This method is particularly useful where the bispecific molecule is a mAb x mAb, mAb x Fab, Fab x F(ab')2 or ligand x Fab fusion protein.
  • a bispecific molecule of the invention can be a single chain molecule comprising one single chain antibody and a binding determinant, or a single chain bispecific molecule comprising two binding determinants. Bispecific molecules may comprise at least two single chain molecules. Methods for preparing bispecific molecules are described or reviewed in, for example in U.S. Patent Number 5,260,203; U.S. Patent Number 5,455,030; U.S.
  • An antibody of the invention further can be prepared using an antibody having one or more of the VH and/or VL sequences disclosed herein as starting material to engineer a modified antibody or antibody "variant", which modified antibody may have altered properties from the parent antibody.
  • An antibody can be engineered by modifying one or more residues within one or both variable regions (i.e. , VH and/or VL), for example within one or more CDR regions and/or within one or more framework regions. Additionally or alternatively, an antibody can be engineered by modifying residues within the constant region(s), for example to alter the effector function(s) of the antibody.
  • other constructs such as antigen-binding fragments, antibody derivatives, immuno- conjugates, and multispecific molecules can be prepared.
  • Standard molecular biology techniques can be used to prepare and express the altered antibody sequence.
  • an antibody variant or derivative typically has at least one altered property as compared to the "parent" antibody
  • the antibody variant or derivative can retain one, some or most of the functional properties of the anti-hNKG2D antibodies described herein, which functional properties include, but are not limited to: (a) prevents NKG2D-mediated activation of an NKG2D-expressing NK or T cell, optionally with an EC50 for reducing ligand-induced cytotoxicity lower than the EC50 for binding to the cell; (b) competes with at least one NKG2D ligand in binding to NKG2D, preferably with at least MICA and ULBP3; (c) reduces the amount of NKG2D on the surface of a NKG2D-expressing NK or T cell, preferably with at least 75%; (d) binds to cynomolgous and/or rhesus NKG2D, preferably with substantially similar efficacy or affinity; (e) binds to more than one form or conformation of NKG2D
  • the functional properties of the antibody variants and derivatives can be assessed using standard assays available in the art and/or described herein.
  • the ability of the antibody to bind hNKG2D can be determined using standard binding assays, such as those set forth in the Examples (e.g., Biacore, flow cytometry, or ELISAs).
  • CDR grafting One type of variable region engineering that can be performed is CDR grafting.
  • Antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain complementarily determining regions (CDRs). For this reason, the amino acid sequences within CDRs are more diverse between individual antibodies than sequences outside of CDRs. Because CDR sequences are responsible for most antibody-antigen interactions, it is possible to express recombinant antibodies that mimic the properties of specific naturally occurring antibodies by constructing expression vectors that include CDR sequences from the specific naturally occurring antibody grafted onto frame- work sequences from a different antibody with different properties (see, e.g., Riechmann, L. et al. (1998) Nature 332:323-327; Jones, P. et al.
  • another embodiment of the invention pertains to an isolated antibody, or antigen binding portion thereof, comprising: a VH region comprising CDR1 , CDR2, and CDR3 sequences comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 15, 21 , 48, and 54, SEQ ID NOs: 16, 22, 49, and 55, and SEQ ID NOs: 17, 23, 50, and 56, respectively, and a VL region comprising CDR1 , CDR2, and CDR3 sequences comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 18, 24, 51 , and 57, SEQ ID NOs: 19, 25, 52, and 58, and SEQ ID NOs: 20, 26, 53, and 59, respectively.
  • such antibodies contain the VH and VL CDR sequences of antibodies 16F16, 16F31 , MS, or 21 F2, yet may contain framework sequences different from
  • the invention also provides a chimeric or humanized version of a murine anti- hNKG2D monoclonal antibody, or antigen-binding fragment thereof, which binds hNKG2D, and the use of such antibodies (e.g., in the modulation of hNKG2D-mediated physiological processes in a mammalian host).
  • the murine antibody is one of ON72, BAT221 , 5C6, 1 D1 1 , 149810, and ECM217.
  • the murine antibody is not one of ON72, BAT221 , 5C6, 1 D1 1 , 149810, and ECM217.
  • such antibodies contain the VH and VL CDR sequences of ON72, BAT221 , 5C6, 1 D1 1 , 149810, or ECM217, or mur- ine monoclonal antibody different from ON72, BAT221 , 5C6, 1 D11 , 149810, ECM217, framework sequences different from these antibodies.
  • the humanized antibody is a humanized version of ON72, comprising e.g. the amino acid sequences of SEQ ID NOS:70 and 71 heavy- and light chain, respectively.
  • Such framework sequences can be obtained from public DNA databases or pub- lished references that include germline antibody gene sequences.
  • germline DNA sequences for human heavy and light chain variable region genes can be found in the "dBase" human germline sequence database (available on the Internet at www.mrc- cpe.cam.ac.uk/vbase), as well as in Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242; Tomlinson, I. M., et al.
  • Preferred framework sequences for use in the antibodies of the invention are those that are structurally similar to the framework sequences used by selected antibodies of the invention, e.g., similar to the VH3_21 , D3-9, JH4, VKIJ.15, and JK2, or VH3_20, D3-10, JH6, VKIII_A27, and JK3, or VH4_59, JH3, VKIII_A27, and JK1 , or VH5_51 , D3_10_R3, JH4, VKIII_L6, and JK1 framework sequences used by the 16F16, 16F31 , MS, and 21 F2 an- tibodies.
  • CDR1 , 2 and 3 sequences of 16F6, 16F31 , MS, or 21 F2 can be grafted onto framework regions that have the same sequence as that found in the germline immunoglobulin gene from which the framework sequence derive, or the CDR sequences can be grafted onto framework regions that contain one or more mutations as compared to the germline sequences. For example, it has been found that in certain instances it is beneficial to mutate residues within the framework regions to maintain or enhance the antigen binding ability of the antibody (see e.g., U.S. Patent Nos. 5,530,101 ; 5,585,089; 5,693,762 and 6,180,370 to Queen et al.).
  • the structural features of anti-hNKG2D antibodies of the invention are used to create structurally related anti-hNKG2D antibodies that retain at least one functional property of the antibodies of the invention, such as binding to hNKG2D.
  • one or more CDR regions of 16F16 or 16F31 can be combined recombinantly with known framework regions and/or other CDRs to create additional, recombinantly-engineered, anti-hNKG2D antibodies of the inven- tion.
  • the starting material for the engineering method is one or more of the VH and/or VL sequences provided herein, or one or more CDR regions thereof.
  • the engineered antibody it is not necessary to actually prepare (i.e., express as a protein) an antibody having one or more of the VH and/or VL sequences provided herein, or one or more CDR regions thereof. Rather, the information contained in the sequence(s) is used as the starting material to create a "second generation" sequence(s) derived from the original sequence(s) and then the "second generation" sequence(s) is prepared and expressed as a protein.
  • the invention provides a method for preparing an anti-hNKG2D antibody comprising: (a) providing: (i) a heavy chain variable region antibody sequence comprising a CDR1 sequence selected from SEQ ID NOs:15, 21 , 48, and 54, a CDR2 sequence selected from SEQ ID NOs:16, 22, 49, and 55, and/or a CDR3 sequence selected from SEQ ID NOs:17, 23, 50, and 56; and (ii) a light chain variable region antibody sequence comprising a CDR1 sequence selected from SEQ ID NOs:18, 24, 51 , and 57, a CDR2 sequence selected from SEQ ID NOs:19, 25, 53, and 59 and/or a CDR3 sequence selected from SEQ ID NOs:20, 26, 53, and 59; (b) altering at least one amino acid residue within the first antibody sequence and/or the second antibody sequence to create at least one altered antibody sequence; and (c) preparing the altered antibody sequence; and (d) expressing the altered
  • variable region modification is to mutate amino acid residues within the VH and/or VL CDR1 , CDR2 and/or CDR3 regions to thereby improve one or more bind- ing properties (e.g. , affinity) of the antibody of interest.
  • Site-directed mutagenesis or PCR- mediated mutagenesis can be performed to introduce the mutation(s) and the effect on antibody binding, or other functional property of interest, can be evaluated in in vitro or in vivo assays as described herein and provided in the Examples.
  • Preferably conservative modifications are introduced.
  • the mutations may be amino acid substitutions, additions or deletions. Moreover, typically no more than 8, more typically no more than 5 residues are altered within a single CDR region.
  • the invention provides isolated anti-hNKG2D antibodies, comprising a heavy chain variable region comprising: (a) a VH CDR1 region comprising an amino acid sequence selected from SEQ ID NOs: 15, 21 , 48, and 54, or an amino acid sequence having one, two, three, four, five, six, seven, or eight amino acid substi- tutions, deletions or additions as compared to an amino acid sequence selected from SEQ ID NOs: 15, 21 , 48, and 54; (b) a VH CDR2 region comprising an amino acid sequence selected from SEQ ID NOs: 16, 22, 49, and 55, or an amino acid sequence having one, two, three, four, five, six, seven or eight amino acid substitutions, deletions or additions as compared to an amino acid sequence selected from SEQ ID NOs: 16, 22, 49, and 55; (c) a VH CDR3 region comprising an amino acid sequence selected from SEQ ID NOs:17, 23, 50, and 56, or an amino acid sequence having one, two, three
  • Engineered antibodies of the invention include those in which modifications have been made to framework residues within VH and/or VL, e.g. to improve the properties of the antibody. Typically such framework modifications are made to decrease the immunogenicity of the antibody. For example, one approach is to "backmutate" one or more framework resi- dues to the corresponding germline sequence. More specifically, an antibody that has undergone somatic mutation may contain framework residues that differ from the germline sequence from which the antibody is derived. Such residues can be identified by comparing the antibody framework sequences to the germline sequences from which the antibody is derived.
  • amino acid residue R1 11 (Kabat residue 103; within FR4) of VH is an arginine whereas this residue in the corresponding germline sequence is a tryptophan (see Figure 5A).
  • some or all of the somatic mutations can be "backmutated" to the germline sequence by, for example, site-directed mutagenesis or PCR-mediated mutagenesis (e.g., residue 11 1 of the VH of 16F16 can be "backmutated” from threonine to alanine).
  • amino acid residue Y95 (within FR3) of the VH region is a tyrisone whereas this residue in the corresponding germline sequence is a histidine (see Figure 5C).
  • the somatic mutation can be "backmutated” from tyrosine to histidine.
  • Kabat residues 3, 6, and 7 of the VH region are histidine (H), aspartic acid (D), and D, respectively, whereas these residues in the corresponding germline sequences are glutamine (Q), glycine (G), and G, respectively (see Figure 5E).
  • framework modification involves mutating one or more residues within the framework region, or even within one or more CDR regions, to remove T cell epi- topes to thereby reduce the potential immunogenicity of the antibody. This approach is also referred to as "deimmunization" and is described in futher detail in U.S. Patent Publication No. 20030153043 by Carr et a/.
  • Fc modifications In addition or as an alternative to modifications made within the framework or CDR regions, antibodies of the invention may be engineered to include modifications within the Fc region, typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, protein stability and/or antigen-dependent cellular cytotoxicity, or lack thereof.
  • an antibody of the invention may be chemically modified (e.g. , one or more chemical moieties can be attached to the antibody) or be modified to alter its glycosylation, again to alter one or more functional properties of the antibody.
  • chemically modified e.g. , one or more chemical moieties can be attached to the antibody
  • modify its glycosylation again to alter one or more functional properties of the antibody.
  • the class of an antibody may be "switched" by known techniques. Such techniques include, e.g., the use of direct recombinant techniques (see e.g., US Patent 4816397) and cell-cell fusion techniques (see e.g., US Patent 5916771 ).
  • direct recombinant techniques see e.g., US Patent 4816397
  • cell-cell fusion techniques see e.g., US Patent 5916771 .
  • an antibody that was originally produced as an IgM molecule may be class switched to an IgG antibody.
  • Class switching techniques also may be used to convert one IgG subclass to another, e.g., from IgGI to lgG2.
  • the effector function of the antibodies of the invention may be changed by isotype switching to, e.g.
  • an IgGI , lgG2, lgG3, lgG4, IgD, IgA, IgE, or IgM antibody for various therapeutic uses.
  • Exemplary cDNA sequences for constant regions are available via, e.g. , GenBank (accessible via NCBI and other public websites), each of which incorporated by reference in its entirety, are as follows:
  • Human IgGI constant heavy chain region GenBank accession No.: J00228;
  • Human lgG2 constant heavy chain region GenBank accession No.: J00230; Human lgG3 constant heavy chain region: GenBank accession No.: X04646;
  • the hinge region of CH1 is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased. This approach is described further in U.S. Patent No. 5677425 by Bodmer et al. The number of cysteine residues in the hinge region of CH1 is altered to, for example, facilitate assembly of the light and heavy chains or to increase or decrease the stability of the antibody.
  • the Fc hinge region of an antibody is mutated to decrease the biological half life of the antibody. More specifically, one or more amino acid mutations are introduced into the CH2-CH3 domain interface region of the Fc-hinge fragment such that the antibody has impaired Staphylococcyl protein A (SpA) binding relative to native Fc-hinge domain SpA binding.
  • SpA Staphylococcyl protein A
  • the antibody is modified to increase its biological half life.
  • one or more of the following mutations can be introduced: T252L, T254S, and T256F, as described in U.S. Patent No. 6277375 to Ward.
  • the antibody can be altered within the CH 1 or CL region to contain a salvage receptor binding epitope taken from two loops of a CH2 domain of an Fc region of an IgG, as described in U.S. Patent Nos. 5869046 and 6121022 by Presta et al.
  • the Fc region is altered by replacing at least one amino acid residue with a different amino acid residue to alter the effecter function ⁇ ) of the antibody.
  • one or more amino acids selected from amino acid residues 234, 235, 236, 237, 297, 318, 320 and 322 can be replaced with a different amino acid residue such that the antibody has an altered affinity for an effector ligand but retains the antigen-binding ability of the parent antibody.
  • the effector ligand to which affinity is altered can be, for example, an Fc receptor or the C1 component of complement. This approach is described in further detail in U.S. Patent Nos. 5624821 and 5648260, both to Winter et al.
  • one or more amino acids selected from amino acid residues 329, 331 and 322 can be replaced with a different amino acid residue such that the antibody has altered C1q binding and/or reduced or abolished complement dependent cytotoxicity (CDC).
  • CDC complement dependent cytotoxicity
  • the Fc region is modified to increase the ability of the antibody to mediate antibody depend- ent cellular cytotoxicity (ADCC) and/or to increase the affinity of the antibody for an Fey receptor by modifying one or more amino acids at the following positions: 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 301 , 303, 305, 307, 309, 312, 315, 320, 322, 324, 326, 327, 329, 330, 331 , 333, 334, 335, 337, 338, 340, 360, 373, 376, 378, 382, 388, 389, 398, 414, 416, 419, 430, 434, 435, 437, 438 or 439.
  • ADCC antibody depend- ent cellular cytotoxicity
  • the constant region may further be modified to stabilize the antibody, e.g., to reduce the risk of a bivalent antibody separating into two monovalent VH-VL fragments.
  • residue S241 may be mutated to a proline (P) residue to al- low complete disulphide bridge formation at the hinge (see, e.g., Angal et al. , MoI Immunol. 1993;30:105-8).
  • the glycosylation of an antibody is modified.
  • an aglycoslated antibody can be made (i.e., the antibody lacks glycosylation).
  • Glycosyla- tion can be altered to, for example, increase the affinity of the antibody for antigen.
  • carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence.
  • one or more amino acid substitutions can be made that result in elimination of one or more variable region framework glycosylation sites to thereby eliminate glycosylation at that site.
  • Such aglycosylation may in- crease the affinity of the antibody for antigen.
  • an antibody can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GlcNac structures.
  • Such altered glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies.
  • carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation "machinery". Cells with such alterations have been described in the art and can be used as host cells in which to express recombinant antibodies of the invention to thereby produce an antibody with altered glycosylation.
  • EP1 176195 by Hanai et al.
  • glyco-protein-modifying glycosyl transferases e.g., beta(l,4)-N-acetylglucosaminyltransferase III (GnTIII)
  • GnTIII glyco-protein-modifying glycosyl transferases
  • mutations can be introduced randomly or selectively along all or part of an anti-hNKG2D antibody coding sequence (e.g., 16F16, 16F31 , MS, or 21 F2 coding sequence) and the resulting modified antibodies can be screened for binding activity and/or other functional properties as described herein.
  • Mutational methods have been described in the art. For example, PCT Publication WO 02/092780 by Short describes methods for creating and screening antibody mutations using saturation mutagenesis, synthetic ligation assembly, or a combination thereof.
  • PCT Publication WO 03/074679 by Lazar et al. describes methods of using computational screening methods to optimize physiochemical properties of antibodies.
  • Antibody derivatives within the scope of this invention include anti-hNKG2D antibodies conjugated or covalently bound to a second agent.
  • the invention provides immunoconjugates comprising an antibody conjugated or covalently bonded to a cytotoxic agent.
  • cytotoxic agent as used herein is a molecule that is capable of killing a cell bearing a hNKG2D receptor on its cell surface.
  • any type of moiety with a cytotoxic or cytoinhibitory effect can be conjugated to the present antibodies to form a cytotoxic conjugate of the present invention and to inhibit or kill specific NK receptor expressing cells, including therapeutic radioisotopes, toxic proteins, toxic small molecules, such as drugs, toxins, immunomodulators, hormones, hormone antagonists, enzymes, oligonucleotides, enzyme inhibitors, therapeutic radionuclides, angio- genesis inhibitors, chemotherapeutic drugs, vinca alkaloids, anthracyclines, epidophyllotox- ins, taxanes, antimetabolites, alkylating agents, antibiotics, COX-2 inhibitors, SN-38, antimitotics, antiangiogenic and apoptotoic agents, particularly doxorubicin, methotrexate, taxol, CPT-1 1 , camptothecans, nitrogen mustards, gemcitabine, alkyl sulfonates, nitrosoureas, tri-
  • a toxin can be of animal, plant, fungal, or microbial origin, or can be created de novo by chemical synthesis.
  • the antibody is derivatized with a radioactive isotope, such as a therapeutic radionuclide or a radionuclide suitable for detection purposes.
  • a radioactive isotope such as a therapeutic radionuclide or a radionuclide suitable for detection purposes.
  • Any of a number of suitable radioactive isotopes can be used, including, but not limited to, 1-131 , In- dium-11 1 , Lutetium-171 , Bismuth-212, Bismuth-213, Astatine-211 , Copper-62, Copper-64, Copper-67, Yttrium-90, lodine-125, lodine-131 , Phosphorus-32, Phosphorus-33, Scandium- 47, Silver-1 11 , Gallium-67, Praseodymium-142, Samarium-153, Terbium-161 , Dysprosium- 166, Holmium-166, Rhenium-186, Rhenium-188, Rhenium-189
  • the radionuclide preferably has a decay energy in the range of 20 to 6,000 keV, preferably in the ranges 60 to 200 keV for an Auger emitter, 100- 2,500 keV for a beta emitter, and 4,000-6,000 keV for an alpha emitter. Also preferred are radionuclides that substantially decay with generation of alpha-particles.
  • the antibody conjugates of the invention can be used to modify a given biological response, where the drug moiety is not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • proteins may include, for example, an enzymatically active toxin, or active fragment thereof, such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor or interferon-y; or, biological re- sponse modifiers such as, for example, lymphokines, interleukin-l (“IL-1"), interleukin-2 (“IL- 2"), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors.
  • IL-1 interleukin-l
  • IL-2 interleukin-2
  • IL-6 inter
  • the second agent can be linked to the antibody directly or indirectly, using any of a large number of available methods.
  • an agent can be attached at the hinge re- gion of the reduced antibody component via disulfide bond formation, using cross-linkers such as N-succinyl 3-(2-pyridyldithio)proprionate (SPDP), or via a carbohydrate moiety in the Fc region of the antibody (see, e.g., Yu et al. (1994) Int. J.
  • the second agent is a detectable moiety, which can be any molecule that can be quantitatively or qualitatively observed or measured.
  • de- tectable markers useful in the conjugated antibodies of this invention are radioisotopes, fluo- rescent dyes, or a member of a complementary binding pair, such as a member of any one of: and antigen/antibody (other than an antibody to NKG2D), lectin/carbohydrate; avidin/biotin; receptor/ligand; or molecularly imprinted polymer/print molecule systems.
  • the second agent may also or alternatively be a polymer, intended to, e.g., increase the circulating half-life of the antibody.
  • exemplary polymers and methods to attach such polymers to peptides are illustrated in, e.g. , U.S. Pat. Nos. 4766106; 4179337; 4495285; and 4609546.
  • Additional illustrative polymers include polyoxyethylated polyols and polyethylene glycol (PEG) moieties.
  • polyethylene glycol is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (C1-C10) alkoxy-or aryloxy-polyethylene glycol or polyethylene glycol-maleimide.
  • a full-length antibody or antibody fragment can be conjugated to one or more PEG molecules with a molecular weight of between about 1 ,000 and about 40,000, such as between about 2000 and about 20,000, e.g. , about 3,000-12,000.
  • the antibody or fragment typically is reacted with polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to the antibody or antibody fragment.
  • PEG polyethylene glycol
  • the pegylation is carried out via an acylation reaction or an alkylation reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer).
  • the antibody to be pegylated is an aglycosylated antibody.
  • Methods for pegylating proteins are known in the art and can be applied to the antibodies of the invention. See for example, EP154316 by Nishimura et al., International patent application PCT/US04/1 1494, and EP401384 by Ishikawa et al.
  • nucleic acid molecules that encode the antibodies of the invention.
  • the nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form.
  • a nucleic acid is "isolated” or “rendered substantially pure” when purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCI banding, column chromatography, agarose gel electrophoresis and others well known in the art. See, F. Ausubel, et al., ed. (1987) Current Protocols in Molecular Biology, Greene Publishing and Wiley Interscience, New York.
  • a nucleic acid of the invention can be, for example, DNA or RNA and may or may not contain intronic sequences.
  • the nucleic acid is a cDNA molecule. While the following paragraphs re- fer to DNA sequences or use thereof, the same methods or principles can generally be applied to mRNA sequences.
  • Nucleic acids of the invention can be obtained using standard molecular biology techniques.
  • hybridomas e.g. , hybridomas prepared from trans- genie mice carrying human immunoglobulin genes as described further below
  • cDNAs encoding the light and heavy chains of the antibody made by the hybridoma can be obtained by standard PCR amplification or cDNA cloning techniques.
  • nucleic acids encoding the antibody can be recovered from the library.
  • Preferred nucleic acids molecules of the invention are those that encode (or comprise a nucleic acid sequence that encodes) the H and L chain sequences of the 16F16, 16F31 , MS, or 21 F2 antibodies of the lgG4 isotype.
  • DNA sequences encoding the 16F16 VH and VL sequences are shown in SEQ ID NOs: 3 and 4, respectively.
  • DNA sequences encoding the 16F31 VH and VL sequences are shown in SEQ ID NOs: 5 and 6, respectively.
  • DNA sequences encoding the MS VH and VL sequences are those that encode for SEQ ID
  • DNA sequences encoding the 21 F2 VH and VL sequences are those that encode for SEQ ID NOS:46 and 47, respectively.
  • VH and VL segments are obtained, these DNA fragments can be further manipulated by standard recombinant DNA techniques, for example to convert the variable region genes to full-length antibody chain genes, to Fab fragment genes or to a scFv gene.
  • a VL- or VH-encoding DNA fragment is operatively linked to another DNA fragment encoding another protein, such as an antibody constant region or a flexible linker.
  • the term "operatively linked”, as used in this context, is intended to mean that the two DNA fragments are joined such that the amino acid sequences encoded by the two DNA fragments remain in-frame.
  • the isolated DNA encoding the VH region can be converted to a full-length heavy chain gene by operatively linking the VH-encoding DNA to another DNA molecule encoding heavy chain constant regions (CH 1 , CH2 and CH3).
  • CH 1 , CH2 and CH3 The sequences of human heavy chain constant region genes are known in the art (see e.g., Kabat, E. A., el al. (1991 ) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification.
  • the heavy chain constant region can be an IgGI , lgG2, lgG3, lgG4, IgA, IgE, IgM or IgD constant region, but most preferably is an lgG4 constant region.
  • the VH-encoding DNA can be opera- tively linked to another DNA molecule encoding only the heavy chain CH 1 constant region.
  • the isolated DNA encoding the VL region can be converted to a full-length light chain gene (as well as a Fab light chain gene) by operatively linking the VL-encoding DNA to another DNA molecule encoding the light chain constant region, CL.
  • the sequences of human light chain constant region genes are known in the art (see e.g., Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U. S. Department of Health and Human Services, NIH Publication No. 91 -3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification.
  • the light chain constant region can be a kappa or lambda constant region, but most preferably is a kappa constant region.
  • the VH-and VL-encoding DNA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly 4 -Ser) 3 , such that the VH and VL sequences can be expressed as a contiguous single- chain protein, with the VL and VH regions joined by the flexible linker (see e.g., Bird et al. (1988) Science 242:423-426; Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883; McCafferty et al. , (1990) Nature 348:552-554).
  • a flexible linker e.g., encoding the amino acid sequence (Gly 4 -Ser) 3
  • Monoclonal antibodies (mAbs) of the present invention can be produced by a variety of techniques, including conventional monoclonal antibody methodology e.g., the standard somatic cell hybridization technique of Kohler and Milstein (1975) Nature 256: 495. Although somatic cell hybridization procedures are preferred, in principle, other techniques for producing monoclonal antibody can be employed e.g., viral or oncogenic transformation of B lymphocytes.
  • hybridomas One preferred animal system for preparing hybridomas is the murine system.
  • Immunization protocols and techniques for isolation of immunized splenocytes for fusion are known in the art, as are fusion partners (e.g., murine myeloma cells) and fusion procedures.
  • Chimeric or humanized antibodies of the present invention can also be prepared based on the sequence of a murine monoclonal antibody using established techniques. For example, DNA encoding the heavy and light chain immunoglobulins can be obtained from the murine hybridoma of interest and engineered to contain non-murine (e.g., human) immunoglobulin sequences using standard molecular biology techniques.
  • the murine variable regions can be linked to human constant regions using methods known in the art (see e.g., U.S. Patent No. 4816567 to Cabilly et al.).
  • the murine CDR regions can be inserted into a human framework using methods known in the art (see e.g., U.S. Patent No. 5225539 to Winter, and U.S. Patent Nos. 5530101 ; 5585089; 5693762 and 6180370 to Queen et al.).
  • the antibodies of the invention are human monoclonal antibodies.
  • Such human monoclonal antibodies directed against hNKG2D can be generated using transgenic or transchromosomic mice carrying parts of the human immune system rather than the mouse system.
  • transgenic and transchromosomic mice include mice referred to herein as HuMAb mice and KM mice, respectively, and are collectively referred to herein as "human Ig mice.”
  • HuMAb mice (Medarex, Inc.) contains human immunoglobulin gene miniloci that encode unrearranged human heavy (p and y) and K light chain immunoglobulin sequences, together with targeted mutations that inactivate the endogenous, u and K chain loci (see e.g., Lonberg, et al.
  • mice exhibit reduced expression of mouse IgM or K, and, in response to immunization, the introduced human heavy and light chain transgenes undergo class switching and somatic mutation to generate high affinity human IgGK monoclonal (Lonberg, N. et al. (1994), supra; reviewed in Lonberg, N. (1994) Handbook of Experimental Pharmacology 113:49-101 ; Lonberg, N. and Huszar, D. (1995) Intern. Rev. Immunol. 13: 65-93, and Harding, F. and Lonberg, N. (1995) Ann. N. Y. Acad. Sci. 764:536-546).
  • human antibodies of the in- vention can be raised using a mouse that carries human immunoglobulin sequences on transgenes and transchomosomes, such as a mouse that carries a human heavy chain transgene and a human light chain transchromosome.
  • KM mice are described in detail in PCT Publication WO 02/43478 to lshida et al.
  • transgenic animal systems expressing human immunoglobulin genes are avail- able in the art and can be used to raise anti-hNKG2D antibodies of the invention.
  • an alternative transgenic system referred to as the Xenomouse (Abgenix, Inc.) can be used; such mice are described in, for example, U.S. Patent Nos. 5939598; 6075181 ; 61 14598; 6150584 and 6162963 to Kucherlapati et al.
  • alternative transchromo- somic animal systems expressing human immunoglobulin genes are available in the art and can be used to raise anti-hNKG2D antibodies of the invention.
  • mice carrying both a human heavy chain transchromosome and a human light chain tranchromosome referred to as "TC mice” can be used; such mice are described in Tomizuka et al. (2000) Proc. Natl. Acad. Sci. USA 97:722-727 ' .
  • cows carrying human heavy and light chain transchromosomes have been described in the art (Kuroiwa et al. (2002) Nature Biotechnol- ogy 20:889-894) and can be used to raise anti-hNKG2D antibodies of the invention.
  • Human monoclonal antibodies of the invention can also be prepared using phage display methods for screening libraries of human immunoglobulin genes.
  • phage display methods for isolating human antibodies are established in the art. See for example: U.S. Patent Nos. 5223409; 5403484; and 5571698 to Ladner et al.; U.S. Patent Nos. 5427908 and 5580717 to Dower et al.; U.S. Patent Nos. 5969108 and 6172197 to McCafferty et al.; and U.S. Patent Nos. 5885793; 6521404; 6544731 ; 6555313; 6582915 and 6593081 to Griffiths et al.
  • Human monoclonal antibodies of the invention can also be prepared using SCID mice into which human immune cells have been reconstituted such that a human antibody response can be generated upon immunization.
  • SCID mice into which human immune cells have been reconstituted such that a human antibody response can be generated upon immunization.
  • Such mice are described in, for example, U.S. Patent Nos. 5476996 and 5698767 to Wilson et al.
  • mice When human Ig mice are used to raise human antibodies of the invention, such mice can be immunized with a purified or enriched preparation of hNKG2D antigen and/or cells expressing hNKG2D, as described by Lonberg, N. et al. (1994) Nature 368(6474): 856- 859; Fishwild, D. et al. (1996) NatureBiotechnolo.gy 14: 845-851 ; and PCT Publication WO 98/24884 and WO 01/14424.
  • the mice will be 6-16 weeks of age upon the first infusion.
  • a purified or enriched preparation (5-50 ⁇ g) of hNKG2D antigen can be used to immunize the human Ig mice intraperitoneal ⁇ .
  • mice can also be immunized with cells expressing hNKG2D, e.g., a human NK or T-cell line, or a mammalian cell expressing recombinant hNKG2D with or without DAP10, to promote immune responses.
  • Example 1 Detailed procedures to generate fully human monoclonal antibodies to hNKG2D are described in Example 1 below.
  • the form and amount of antigen administered e.g., hNKG2D polypeptide or cell expressing hNKG2D
  • adjuvants such as, e.g., complete Freund's adjuvant or incomplete Freund's adju- vant, are typically optimized for each antigen-mouse system according to established methods in the art.
  • the immune response can be monitored over the course of the immunization protocol with plasma samples being obtained by retroorbital bleeds, and the plasma or serum can be screened by ELISA (as described below), and mice with sufficient titers of anti-hNKG2D human immunoglobulin can be used for fusions. Mice can be boosted intravenously with antigen 3 days before sacrifice and removal of the spleen. It is expected that 2-3 fusions for each immunization may need to be performed.
  • splenocytes and/or lymph node cells from immunized mice can be isolated and fused to an appropriate immortalized cell line, such as a mouse myeloma cell line.
  • an appropriate immortalized cell line such as a mouse myeloma cell line.
  • the resulting hybridomas can be screened for the production of antigen-specific antibodies.
  • single cell suspensions of splenic lymphocytes from immunized mice can be fused to one-sixth the number of P3X63-Ag8.653 nonsecreting mouse myeloma cells (ATCC, CRL 1580) with 50% PEG.
  • the cells can be fused by electrofusion.
  • Cells are plated at approximately 2 x 10 5 in a flat bottom microtiter plate, followed by a two week incubation in selective medium containing 20% fetal Clone Serum, 18% "653" conditioned media, 5% origen (IGEN), 4 mM L-glutamine, 1 mM sodium pyruvate, 5mM HEPES, 0.055 mM 2-mercaptoethanol, 50 units/ml penicillin, 50 mg/ml streptomycin, 50 mg/ml gentamycin and 1X HAT (Sigma; the HAT is added 24 hours after the fusion). After approximately two weeks, cells can be cultured in medium in which the HAT is replaced with HT.
  • selective medium containing 20% fetal Clone Serum, 18% "653" conditioned media, 5% origen (IGEN), 4 mM L-glutamine, 1 mM sodium pyruvate, 5mM HEPES, 0.055 mM 2-mercaptoethanol, 50 units/ml pen
  • Supernatants can be filtered and concentrated before affinity chromatography with protein A-sepharose (Pharmacia, Piscata- way, N. J.). Eluted IgG can be checked by gel electrophoresis and high performance liquid chromatography to ensure purity. The buffer solution can be exchanged into PBS, and the concentration can be determined by spectroscopy. The monoclonal antibodies can be ali- quoted and stored at -80° Antibodies of the invention can also be produced in a host cell transfectoma using, for example, a combination of recombinant DNA techniques and gene transfection methods as is well known in the art (e.g., Morrison, S. (1985) Science 229:1202).
  • DNAs encoding partial or full-length light and heavy chains can be obtained by standard molecular biology techniques (e.g. PCR amplification or cDNA cloning using a hybridoma that expresses the antibody of interest) and the DNAs can be inserted into expression vectors such that the genes are operatively linked to transcriptional and translational control sequences and may serve their intended function of regulating the transcription and translation of the antibody gene.
  • the expression vector and expression control sequences are chosen to be compatible with the expression host cell used.
  • the antibody light chain gene and the antibody heavy chain gene can be inserted into separate vector or, more typically, both genes are inserted into the same expression vector.
  • the antibody genes are inserted into the expression vector by standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present).
  • the light and heavy chain variable regions of the antibodies described herein can be used to create full- length antibody genes of any antibody isotype by inserting them into expression vectors already encoding heavy chain constant and light chain constant regions of the desired isotype such that the VH segment is operatively linked to the CH segment(s) within the vector and the VL segment is operatively linked to the CL segment within the vector.
  • the recombinant expression vector can encode a signal peptide that facilitates secretion of the antibody chain from a host cell.
  • the antibody chain gene can be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the antibody chain gene.
  • the signal peptide can be an immunoglobulin signal peptide or a heterolo- gous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein).
  • the recombinant expression vectors of the invention carry regulatory sequences that control the expression of the antibody chain genes in a host cell.
  • the term "regulatory sequence” is intended to include promoters, enhancers and other expression control elements (e.g. polyadenylation signals) that control the tran- scription or translation of the antibody chain genes.
  • Such regulatory sequences are described, for example, in Goeddel (Gene Expression Technology. Methods in Enzymology 185, Academic Press, San Diego, CA (1990)).
  • the design of the expression vector may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc.
  • Pre- ferred regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV), Simian Virus 40 (SV40), adenovirus, (e.g., the adenovirus major late promoter (AdMLP) and polyoma.
  • CMV cytomegalovirus
  • SV40 Simian Virus 40
  • AdMLP adenovirus major late promoter
  • nonviral regulatory se- quences may be used, such as the ubiquitin promoter or p-globin promoter.
  • regulatory elements composed of sequences from different sources, such as the SRa promoter system, which contains sequences from the SV40 early promoter and the long terminal repeat of human T cell leukemia virus type 1 (Takebe, Y. et al. (1988) MoI. Cell. Biol. 8:466-472).
  • the recombinant expression vectors of the invention may carry additional sequences, such as sequences that regulate replication of the vector in host cells (e.g. origins of replication) and selectable marker genes.
  • the selectable marker gene facilitates selection of host cells into which the vector has been introduced (see, e.g. U.S. Pat. Nos.
  • the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced.
  • Preferred selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).
  • DHFR dihydrofolate reductase
  • the expression vector(s) encoding the heavy and light chains is transfected into a host cell by standard techniques.
  • transfection are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like.
  • prokaryotic or eukaryotic host cells expression of antibodies in eukaryotic cells, and most preferably mammalian host cells, is the most preferred because such eukaryotic cells, and in particular mammalian cells, are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody.
  • Prokaryotic expression of anti- body genes has been reported to be ineffective for production of high yields of active antibody (Boss, M. A. and Wood, C. R. (1985) Immunology Today 6:12-13).
  • Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr-CHO cells, described in Urlaub and Chasin, (1980) Proc. Nail. Acad. Sci. USA 77:4216-4220, used with a DHFR se- lectable marker, e. g., as described in R. J. Kaufman and P. A. Sharp (1982) MoI. Biol. 159:601 -621), NSO myeloma cells, COS cells and SP2 cells.
  • Chinese Hamster Ovary CHO cells
  • dhfr-CHO cells described in Urlaub and Chasin, (1980) Proc. Nail. Acad. Sci. USA 77:4216-4220, used with a DHFR se- lectable marker, e. g., as described in R. J. Kaufman and P. A. Sharp (1982) MoI. Biol. 159:601 -6
  • another preferred expression system is the GS gene expression system disclosed in WO 87/04462, WO 89/01036 and EP 338,841.
  • the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown.
  • Antibodies can be recovered from the culture medium using standard protein purification methods.
  • Antibody characterization After production or purification, or as part of a screening or selection procedure, the functional characteristics of an anti-hNKG2D antibody of the invention can be investigated.
  • Functional properties of interest include, e.g., antibody binding specificity for hNKG2D, antibody competition with hNKG2D-ligands, antibody competition with reference antibodies (such as, e.g., 16F16, 16F31 , MS, and 21 F2), the epitope to which the antibody binds, the affinity of the antibody-antigen interaction, and antagonistic/agonistic properties of the antibody.
  • Antibody specificity for hNKG2D can be evaluated by confirming that the mono- clonal antibody (or, as part of animal screening procedures, serum containing polyclonal ant- bodies) binds NKG2D expressing cells but not NKG2D negative cells. Cell lines with or without NKG2D are incubated with antibody followed by incubation with secondary antibody directly labelled, and visualised by, e.g., flow cytometry.
  • Blockade of ligand binding can be evaluated by incubating cells expressing NKG2D with or without antibody or hybridoma supernatant, followed by incubation with a
  • Ngand-mFc protein and a secondary antibody specific for the ligand and the level of ligand binding and blockade thereof determined by flow cytometry.
  • Blockade can be calculated as the % ligand binding with pre-incubation compared to without pre-incubation, when lower binding is seen upon pre-incubation.
  • Competition for binding site used by one or more reference anti-NKG2D antibodies can be evaluated in a similar manner, except that the pre-incubation can either performed with an antibody of the invention or the reference antibody (e.g., ON72 or 149810), followed by incubation with and detection of the subsequently added antibody.
  • Affinity parameters, including on- and off- rate, of antibodies can determined on a Biacore machine.
  • hNKG2D-Fc protein can be immobilized on a chip, the antibody passed over the chip, the on- and off-rates determined, and the KD calculated.
  • Induction of NKG2D internalisation by antibodies can be measured by incubati- ing hNKG2D-expressing cells with or without antibody overnight, followed by re-addition of the antibody and detection of the level of NKG2D (i.e. the level of antibody bound) in a flow cytometer.
  • hNKG2D-ligand mediated killing can be assessed, using, e.g., the NK cell lines NK92 or NKL as effector cells that kill 51 Cr-loaded target cells expressing NKG2D ligand, either MICA, MICB, or ULBP1 -4.
  • Activation of NKG2D upon antibody binding can be measured as induction of cell-proliferation of CD8+ cells in a PBMC population upon stimulation via the T-cell receptor, CD28 and or NKG2D, with or without pre-stimulation (e.g. , via TCR, CD28 and IL-2 or IL-15).
  • Binding Assays The present invention provides for antibodies, and antigen-binding fragments and immunoconjugates thereof, that bind hNKG2D. Any of a wide variety of assays can be used to assess binding of an antibody to hNKG2D. Protocols based upon ELISAs, radioimmunoassays, Western blotting, BIACORE, and other competition assays, inter alia, are suitable for use and are well known in the art. Further, several binding assays, including competition as- says, are described in the Examples.
  • simple binding assays can be used, in which a test antibody is incubated in the presence of a target protein or epitope (e.g., NKG2D or a portion thereof), unbound antibodies are washed off, and the presence of bound antibodies is assessed using, e.g., radiolabels, physical methods such as mass spectrometry, or direct or indirect fluores- cent labels detected using, e.g., cytofluorometric analysis (e.g. FACScan). Such methods are well known to those of skill in the art. Any amount of binding above the amount seen with a control, non-specific antibody indicates that the antibody binds specifically to the target.
  • a target protein or epitope e.g., NKG2D or a portion thereof
  • unbound antibodies are washed off
  • the presence of bound antibodies is assessed using, e.g., radiolabels, physical methods such as mass spectrometry, or direct or indirect fluores- cent labels detected using, e.g., cytofluorometric
  • the ability of the test antibody to bind to the target cell or human NKG2D can be compared with the ability of a (negative) control protein, e.g. an antibody raised against a structurally unrelated antigen, or a non-lg peptide or protein, to bind to the same target.
  • a (negative) control protein e.g. an antibody raised against a structurally unrelated antigen, or a non-lg peptide or protein
  • Antibodies or fragments that bind to the target cells or NKG2D using any suitable assay with 25%, 50%, 100%, 200%, 1000%, or higher increased affinity relative to the control protein, are said to "specifically bind to" or "specifically interact with” the target, and are preferred for use in the therapeutic methods described below.
  • the ability of a test anti- body to affect the binding of a (positive) control antibody against NKG2D e.g. 16F16, 16F31 , MS, or 21 F2, may also be assessed.
  • the invention provides for anti-hNKG2D antibodies sharing biological characteristics and/or substantial VH and/or VL sequence identity with 16F16, 16F31 , MS, or 21 F2.
  • One exemplary biological characteristic is the binding to the 16F16, 16F31 , MS, or 21 F2 epitope, i.e. , the respective regions in the extracellular domain of hNKG2D to which the 16F16, 16F31 , MS, or 21 F2 antibodies bind.
  • a routine cross-blocking assay such as that described in Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory, Ed Harlow and David Lane (1988), can be performed.
  • control antibody and a test antibody are admixed (or pre-adsorbed) and applied to a sample containing NKG2D.
  • control antibodies e.g., 1 :10 or 1 :100
  • the control and varying amounts of test antibody can simply be admixed during exposure to the antigen/target sample.
  • the control antibody As long as one can distinguish bound from free antibodies (e.g., by using separation or washing techniques to eliminate unbound antibodies) and the control antibody from test antibody (e.g., by using species- or isotype-specific secondary antibodies, by specifically labeling the control antibody with a detectable label, or by using physical methods such as mass spec- trometry to distinguish between different compounds) one will be able to determine if the test antibody reduces the binding of the control antibody to the antigen, indicating that the test antibody recognizes substantially the same epitope as the control. In this assay, the binding of the (labeled) control antibody in the presence of a completely irrelevant antibody is the control high value. The control low value is be obtained by incubating the labeled (positive) control antibody with unlabeled control antibody, where competition would occur and reduce binding of the labeled antibody.
  • a significant reduction in labeled antibody reactivity in the presence of a test antibody is indicative of a test antibody that recognizes the same epitope, i.e., one that "cross-reacts" with the labeled control antibody.
  • Any test antibody or compound that re- Jerusalem the binding of the labeled control to the antigen/target by at least 50% or more pref- erably 70%, at any ratio of control:test antibody or compound between about 1 :10 and about 1 : 100 is considered to be an antibody or compound that binds to substantially the same epitope or determinant as the control.
  • such test antibody or compound will reduce the binding of the control to the antigen/target by at least 90%. Nevertheless, any compound or antibody that reduces the binding of a control antibody or compound to any measurable extent can be used in the present invention.
  • competition can be assessed by a flow cytometry test.
  • Cells bearing hNKG2D are incubated first with a control antibody that is known to specifically bind to the receptor (e.g. , T or NK cells expressing hNKG2D or BaF/3 cell recombinant express- ing hNKG2D, and 16F16, 16F31 , MS, or 21 F2 antibody), and then with the test antibody that may be labeled with, e.g., a fluorochrome or biotin.
  • test antibody is said to compete with the control if the binding obtained with preincubation with saturating amounts of control antibody is 80%, preferably, 50%, 40% or less of the binding (mean of fluorescence) obtained by the antibody without preincubation with the control.
  • a test antibody is said to compete with the control if the binding obtained with a labeled control (by a fluorochrome or biotin) on cells preincubated with saturating amount of antibody to test is 80%, preferably 50%, 40%, or less of the binding obtained without preincubation with the antibody. See Example 5 for an exemplary antibody competition assay.
  • Similar cross-blocking assays can also be used to evaluate whether a test (human- ized) antibody affects the binding of a natural ligand for human NKG2D, such as MICA, MICB, ULBP1 , ULBP2, ULBP3, ULBP4, or a member of the RAET1 family, simply by exchanging 16F16, 16F31 , MS, or 21 F2 for a suitable form of the hNKG2D-ligand.
  • a suitable form described in the Examples, are fusion proteins of the ligand (e.g., MICA) with the Fc-portion of an antibody.
  • Having the ligand conjugated to an Fc-region allows for detection of the fusion protein by antibodies specific for the animal species from which the Fc-region derives, using, e.g., goat-anti-mouse antibodies to detect a murine Fc-region.
  • a cellular assay is used in which hNKG2D-expressing cells, e.g., CD4 + CD28 cells from rheumatoid arthritis patients (or the equivalent cells from another autoimmune or inflammatory disorder) are incubated with an NKG2D ligand such as MICA, MICB, or a ULBP protein, e.g., in the form of an Fc-fusion protein, or a cell expressing any of these ligands, and the ability of an anti-NKG2D antibody or other molecule to block the activation of the cell is assessed.
  • an NKG2D ligand such as MICA, MICB, or a ULBP protein
  • a baseline level of activity for the NKG2D receptor is obtained in the absence of a ligand, and the ability of the antibody or compound to cause a decrease in the baseline activity level is detected.
  • a high-throughput screening approach is used to identify compounds capable of blocking the activation of the receptor, or otherwise downregulating it. See Example 3 for an exemplary ligand competition assay.
  • monoclonal antibodies that recognize an NKG2D epitope will react with an epitope that is present on a substantial percentage of CD4+ T cells, particularly CD4+CD28- T cells, in patients such as rheumatoid arthritis patients, but will not significantly react with other cells, i.e., immune or non-immune cells that do not express NKG2D. Accordingly, once an antibody that specifically recognizes hNKG2D on NK or T cells, it can be tested for its ability to bind to T cells taken from patients with autoimmune or inflammatory disorders such as rheumatoid arthritis.
  • the present invention can be used for the treatment of any disorder in which NKG2D activity is linked to the pathology of the disorder, regardless of the cell type expressing the receptor (e.g., CD4+ T cells, CD8+ T cells, NK cells, etc.), and the antibodies can be tested for their ability to bind to the receptor on whichever cell type is relevant for the particular disorder. For example, if it is observed that a particular disorder is associated with excess activity or proliferation of NKG2D- expressing NK cells, then the antibodies can be developed and tested using NK cells expressing the same receptor.
  • the cell type expressing the receptor e.g., CD4+ T cells, CD8+ T cells, NK cells, etc.
  • the antibodies are validated in an immunoassay to test its ability to bind to NKG2D-expressing cells, e.g. CD4+CD28- T cells taken from patients with rheumatoid arthritis.
  • NKG2D-expressing cells e.g. CD4+CD28- T cells taken from patients with rheumatoid arthritis.
  • peripheral blood lymphocytes PBLs
  • CD4+, preferably CD4+ CD28-, cells are enriched from the PBLs, e.g., by flow cytometry using relevant antibodies.
  • the ability of a given antibody to bind to the cells is then assessed using standard methods well known to those in the art.
  • a substantial proportion e.g. 20%, 30%, 40%, 50%, 60%, 70%, 80% or more
  • NK cells e.g. NK cells, CD8 T cells, CD4 T cells from RA patients, etc.
  • a significant percentage of patients e.g., 5%, 10%, 20%, 30%, 40%, 50%
  • the antibod- ies can either be directly or indirectly labeled.
  • indirectly labeled a secondary, labeled antibody is typically added.
  • the binding of the antibodies to the cells can then be detected using, e.g., cytofluorometric analysis (e.g. FACS).
  • FACS cytofluorometric analysis
  • the antibodies of the invention preferably do not demonstrate substantial specific binding to Fc receptors.
  • Such antibodies may comprise constant regions of various heavy chains that are known not to bind Fc receptors.
  • One such example is an lgG4 constant region.
  • antibody fragments that do not comprise contstant regions, such as Fab or F(ab')2 fragments, can be used to avoid Fc receptor binding.
  • Fc receptor binding can be assessed according to methods known in the art, including for example testing bind- ing of an antibody to Fc receptor protein in a BIACORE assay.
  • any other antibody type can be used in which the Fc portion is modified to minimize or eliminate binding to Fc receptors (see, e.g. , WO03101485, the disclosure of which is herein incorporated by referece).
  • Assays such as, e.g., cell based assays, to assess Fc receptor binding are well known in the art, and are described in, e.g. , WO03101485. Functional assays
  • any suitable physiological change that reflects NKG2D activity can be used to assess the utility of a test compound or antibody.
  • the activity of the receptor can be assessed by detecting the expression of NKG2D-responsive genes, e.g., CD25, IFN-gamma, or TNF-alpha (see, e.g., Groh et al.
  • NKG2D activity can be assessed by incubating CD4+CD28-NKG2D+ cells in the presence of a ligand or activating anti-NKG2D antibody, as well as an anti-CD3 antibody, to evaluate the ability of the compound or test antibody to inhibit the release of TNF-alpha or IFN-gamma by the T cells.
  • CD4+CD28-NKG2D+ T cells can be incubated in the presence of ligand, e.g., MICA, MICB, ULBP-1 , ULBP-2, ULBP-3, etc., or ligand-producing cells, e.g., autologous MIC+ RA synoviocytes, and the ability of the test an- tibody or compound to inhibit cytokine production (e.g., IFN-gamma or TNF-alpha), or T cell proliferation assessed.
  • ligand e.g., MICA, MICB, ULBP-1 , ULBP-2, ULBP-3, etc.
  • ligand-producing cells e.g., autologous MIC+ RA synoviocytes
  • cytokine production e.g., IFN-gamma or TNF-alpha
  • In vitro assays can optionally use cells taken from patients with autoimmune or inflammatory disorders such as RA, e.g. CD4+CD28- cells expressing NKG2D taken from (or cell lines derived therefrom) patients with RA, but in general any NKG2D-expressing cells can be used.
  • RA autoimmune or inflammatory disorders
  • non-RA immune cell lines e.g. T cell lines
  • Cell lines can, for example, be established using CD4+CD28- NKG2D+ cells from RA patients, e.g. PBLs or T cells isolated from synovial tissue. Such cells can be cultured in the presence of IL-15 to ensure continued expression of NKG2D (see, e. g., Groh et al. (2003) PNAS 100: 9452-9457, the entire disclosure of which is herein incorporated by reference).
  • an anti-hNKG2D antibody reduces or blocks NKG2D interactions with one or more of its ligands, or competes with an antibody known to block hNKG2D ligand interaction, it can be useful for reducing NKG2D-mediated activation of NK or T cells. This can be evaluated by a typical cytotoxicity assays.
  • Example 6 describes an exemplary cytotoxicity assay, NKG2D-ligand mediated killing of target cells.
  • the ability of anti-hNKG2D antibodies to reduce or inhibit the NK cell-mediated killing of MICA-transfected BaF/3 is assessed by measuring target cell release of 51Cr.
  • assays can be used for this purpose, including the following.
  • One assay can evaluate proliferation and cytokine production after activation with antibodies, either soluble or plate-bound, in combination with anti-CD3 and/or anti-CD28 an- tibodies, of PBMCs from healthy volunteers or IBD patients.
  • PBMCs are purified by conventional methods from healthy subjects or inflammatory bowel disease (IBD) patients.
  • the cells are stained with CFSE (from Molecular probes, cat #C34554).
  • To 10 7 cells in 0.5 ml PBS with 2% FCS
  • 1 ⁇ l CFSE 0.5 mM
  • FCS 2 ml FCS is added, and the mixture is left for 1 min at room tempera- ture.
  • the cells are then washed 3 times by centrifugation with RPMI-1640 medium (12 ml). After wash, the cells are resuspended in 1 ml media (e.g. RPMI-1640) with 2% FCS.
  • PBMCs 100.000 CFSE-labelled PBMCs are added and left for 3 days.
  • Supernatant is then collected for analysis of cytokines, and the PBMCs are analysed by flow cytometry with regard to the type of lymphocyte with anti-CD56, anti-CD4, anti-CD8, and CFSE labelling for proliferation.
  • the effect on the cytotoxic potential of CD8+ T cells towards a target cells lacking NKG2D ligands is tested. If binding boosts the cytotoxic potential of the cells, agonistic activity is present.
  • IL-2 stimulated PBMC from healthy subjects are incubated with p815 cells expressing MICA, or with untransfected p815 cells and an anti- CD3 antibody, (which will lead to redirected killing by binding to the Fc receptors on p815 cells) and CD8 cytotoxic T cells. It is then analyzed whether an anti-NKG2D antibody that does not bind to p815 cells (e.g., an antibody of human lgG4 isotype) blocks MICA-NKG2D- directed binding and/or if the antibody boosts CD3-p815 redirected binding. In this manner, it can be shown that the activity of the CD8+ T cells is not enhanced by incubating p815 cells with an anti-CD3 antibody and an additional anti-NKG2D antibody, while the same anti-
  • NKG2D antibody can shown to be functional by demonstrating that it blocks NK-MICA interaction on p815-induced killing in the same PBMC population.
  • NK cell lines such as, e.g. , NKL cells or NK-92 cells
  • human NK or CD8+ T cells isolated from peripheral blood can be used.
  • NKL cells can be incubated with a human anti-NKG2D antibody in solution or plate bound, with, e.g., Fc-MICA or irradiated MICA expressing cells as a control.
  • the cells After incubation for suitable time periods, (e.g., 5, 10, 30 min), the cells are lysed in the presence of protease and phosphatase inhibitors on ice, and analyzed for the levels of one or more phosphorylated signalling molecules that are known to be downstream of stimulation of NKG2D (e.g., Pi3K, Akt, and vav), by standard Western blotting techniques.
  • NKG2D e.g., Pi3K, Akt, and vav
  • any physiological or pathological consequence of NKG2D activation in cells within the animal can be used to assess antibody or test compound activity.
  • CD4+CD28-NKG2D+ cells can be introduced into the joints of an ani- mal model, with or without co-administration of ligand producing cells such as MICA- producing synoviocytes, and inflammation or tissue damage is assessed. Test compounds or antibodies can then be introduced, and their ability to inhibit, slow, reverse, or in any way affect the inflammation or tissue damage is detected.
  • ligand producing cells such as MICA- producing synoviocytes
  • rheumatoid arthritis (RA) synovial explants can also be performed to study the effects of blocking NKG2D on spontaneous release of pro-inflammatory cytokines (see, e.g.,Brennan et al., Lancet 1989; 2 (8657);244-247).
  • human or humanized anti-hNKG2D antibodies are tested on RA synovial membrane cultures and compared to, e.g., murine anti-hNKG2D antibodies at concentrations shown to be useful to block ligand binding and function of NKG2D.
  • RA synovial cells are cultured for 48 hrs in the ab- sence or presence of anti-NKG2D antibodies or an isotype control antibody.
  • Known anti- inflammatory drugs can be used as positive controls.
  • the effects of the anti-NKG2D antibodies are initially tested at concentrations up to 30 ⁇ g/ml on 6 RA synovial membranes. Viability of the cells is analysed in a assay staining living cells (e.g. a MTT assay) to determine if the added reagent has any cytotoxicity.
  • ELISA is then used to detect cytokines such as, e.g., TNF ⁇ , IL-1 ⁇ and IL-6 levels in culture supernatants.
  • antibodies of the invention can be tested in experimental models of, e.g., psoriasis or ulcerative colitis.
  • Psoriasis-affected skin sample can be transplanted onto a SCID mouse together with the patients own PBMCs, and the effect of introduction of a test compound and their ability to inhibit, slow, reverse, or in any way affect the inflammation or tissue damage, can be detected.
  • Kjellev et al. (Eur J Immunol 2008;37: 1397-1406) and lto et al. (Am J Physiol Gastrointest Liver Physiol 2008;294:G199-G207) describe experimental models for assessing treatment of ulcerative colitis using anti-murine NKG2D antibody.
  • the present invention provides a pharmaceutical composition or formulation comprising anti-hNKG2D antibodies as described herein together with one or more carriers.
  • one exemplary aspect of the invention is a pharmaceutical formulation comprising such an antibody which is present in a concentration from 1 mg/ml to 500 mg/ml, and wherein said formulation has a pH from 2.0 to 10.0.
  • the formulation may further com- prise a buffer system, preservative(s), tonicity agent(s), chelating agent(s), stabilizers, and/or surfactants.
  • the pharmaceutical formulation is an aqueous formulation, i.e., formulation comprising water. Such formulation is typically a solution or a suspension.
  • the pharmaceutical formulation is an aqueous solution.
  • aqueous formulation is defined as a formulation comprising at least 50 %w/w water.
  • the term "aqueous solution” is defined as a solution comprising at least 50 %w/w water
  • aqueous suspension is defined as a suspension comprising at least 50 %w/w water.
  • the pharmaceutical formulation is a freeze-dried formulation, whereto the physician or the patient may add solvents and/or diluents prior to admini- stration.
  • the pharmaceutical formulation is a dried formulation (e.g. freeze-dried or spray-dried) ready for use without any prior dissolution.
  • the pharmaceutical formulation comprises an aqueous solution of such an antibody, and a buffer, wherein the anitbody is present in a concentration from 1 mg/ml or above, and wherein said formulation has a pH from about 2.0 to about 10.0.
  • the pH of the formulation is in the range selected from the list consisting of from about 2.0 to about 10.0, about 3.0 to about 9.0, about 4.0 to about 8.5, about 5.0 to about 8.0, and about 5.5 to about 7.5.
  • the formulation includes a buffer that is selected from the group consisting of sodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, arginine, sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium phosphate, and tris(hydroxymethyl)-aminomethan, bicine, tricine, malic acid, succinate, maleic acid, fumaric acid, tartaric acid, aspartic acid or mixtures thereof.
  • a buffer that is selected from the group consisting of sodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, arginine, sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium phosphate, and tris(hydroxymethyl)-aminomethan, bicine, tricine, malic acid, succinate, maleic acid, fumaric acid, tartaric acid, aspartic acid or mixtures thereof.
  • the formulation also or alternatively comprises a pharmaceutically acceptable preservative.
  • the preservative may be selected from, e.g., the group consisting of phenol, o-cresol, m-cresol, p-cresol, methyl p-hydroxybenzoate, propyl p- hydroxybenzoate, 2-phenoxyethanol, butyl p-hydroxybenzoate, 2-phenylethanol, benzyl alcohol, chlorobutanol, and thiomerosal, bronopol, benzoic acid, imidurea, chlorohexidine, sodium dehydroacetate, chlorocresol, ethyl p-hydroxybenzoate, benzethonium chloride, chlor- phenesine (3p-chlorphenoxypropane-1 ,2-diol) or mixtures thereof.
  • the preservative may, e.g., be present in a concentration from 0.1 mg/ml to 20 mg/ml, from 0.1 mg/ml to 5 mg/ml, from 5 mg/ml to 10 mg/ml, or from 10 mg/ml to 20 mg/ml.
  • concentration from 0.1 mg/ml to 20 mg/ml, from 0.1 mg/ml to 5 mg/ml, from 5 mg/ml to 10 mg/ml, or from 10 mg/ml to 20 mg/ml.
  • Each one of these specific preservatives constitutes an alternative embodiment of the invention.
  • the use of a preservative in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington: The Science and Practice of Pharmacy, 19 th edition, 1995.
  • the formulation also or alternatively comprises an isotonic agent.
  • the isotonic agent may be, e.g., selected from the group consisting of a salt (e.g.
  • sodium chloride a sugar or sugar alcohol
  • an amino acid e.g. L-glycine, L-histidine, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine
  • an alditol e.g. glycerol (glycerine), 1 ,2-propanediol (propyleneglycol), 1 ,3-propanediol, 1 ,3-butanediol
  • polyethyleneglycol e.g. PEG400
  • Any sugar such as mono-, di-, or polysaccharides, or water- soluble glucans, including for example fructose, glucose, mannose, sorbose, xylose, maltose, lactose, sucrose, trehalose, dextran, pullulan, dextrin, cyclodextrin, soluble starch, hy- droxyethyl starch and carboxymethylcellulose-Na may be used.
  • the sugar additive is sucrose.
  • Sugar alcohol is defined as a C4-C8 hydrocarbon having at least one -OH group and includes, for example, mannitol, sorbitol, inositol, galactitol, dulcitol, xyli- tol, and arabitol.
  • the sugar alcohol additive is mannitol.
  • the sugars or sugar alcohols mentioned above may be used individually or in combination. There is no fixed limit to the amount used, as long as the sugar or sugar alcohol is soluble in the liquid preparation and does not adversely effect the stabilizing effects achieved using the methods of the invention.
  • the sugar or sugar alcohol concentration can, e.g., be between about 1 mg/ml and about 150 mg/ml.
  • the isotonic agent can be present in a concentration from, e.g., 1 mg/ml to 50 mg/ml, from 1 mg/ml to 7 mg/ml, from 8 mg/ml to 24 mg/ml, or from 25 mg/ml to 50 mg/ml.
  • concentration from, e.g., 1 mg/ml to 50 mg/ml, from 1 mg/ml to 7 mg/ml, from 8 mg/ml to 24 mg/ml, or from 25 mg/ml to 50 mg/ml.
  • the formulation also or alternatively comprises a chelating agent.
  • the chelating agent can, for example, be selected from salts of ethylenediamine- tetraacetic acid (EDTA), citric acid, and aspartic acid, and mixtures thereof.
  • the chelating agent may, for example, be present in a concentration from 0.1 mg/ml to 5 mg/ml, from 0.1 mg/ml to 2 mg/ml, or from 2 mg/ml to 5 mg/ml.
  • EDTA ethylenediamine- tetraacetic acid
  • the chelating agent may, for example, be present in a concentration from 0.1 mg/ml to 5 mg/ml, from 0.1 mg/ml to 2 mg/ml, or from 2 mg/ml to 5 mg/ml.
  • Each one of these specific chelating agents constitutes an alternative embodiment of the invention.
  • the use of a chelating agent in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington
  • compositions of the invention can be stabilized liquid pharmaceutical compositions whose therapeutically active components in- elude a polypeptide that possibly exhibits aggregate formation during storage in liquid pharmaceutical formulations.
  • aggregate formation is intended a physical interaction between the polypeptide molecules that results in formation of oligomers, which may remain soluble, or large visible aggregates that precipitate from the solution.
  • uring storage is intended a liquid pharmaceutical composition or formulation once prepared, is not immediately admin- istered to a subject.
  • liquid pharmaceutical composition or formulation is dried either by freeze drying (i.e., lyophilization; see, for example, Williams and PoIIi (1984) J. Parenteral Sci. Technol. 38:48-59), spray drying (see Masters (1991) in Spray-Drying Handbook (5th ed; Longman Scientific and Technical, Essez, U.K.), PP- 491-676; Broadhead et al. (1992) Drug Devel. Ind. Pharm. 18:1169-1206; and Mumenthaler et al.
  • compositions of the invention may alternatively or further com- prise an amount of an amino acid base sufficient to decrease aggregate formation by the polypeptide during storage of the composition.
  • amino acid base is intended an amino acid or a combination of amino acids, where any given amino acid is present either in its free base form or in its salt form. Where a combination of amino acids is used, all of the amino acids may be present in their free base forms, all may be present in their salt forms, or some may be present in their free base forms while others are present in their salt forms.
  • amino acids to use in preparing the compositions of the invention are those carrying a charged side chain, such as arginine, lysine, aspartic acid, and glutamic acid.
  • Any stereoisomer i.e., L, D, or a mixture thereof
  • a particular amino acid e.g. methionine, his- tidine, imidazole, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine and mix- tures thereof
  • a particular amino acid e.g. methionine, his- tidine, imidazole, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine and mix- tures thereof
  • the L-stereoisomer is used.
  • Compositions of the invention may also be formulated with analogues of these amino acids.
  • amino acid analogue is intended a derivative of the naturally occurring amino acid that brings about the desired effect of decreasing aggregate formation by the polypeptide during storage of the liquid pharmaceutical compositions of the invention.
  • Suitable arginine analogues include, for example, aminoguanidine, ornithine and N-monoethyl L-arginine
  • suitable methionine analogues include ethionine and buthionine
  • suitable cysteine analogues include S-methyl-L cysteine.
  • the amino acid analogues are incorporated into the compositions in either their free base form or their salt form.
  • the amino acids or amino acid analogues are used in a concentration, which is sufficient to prevent or delay aggregation of the protein.
  • methionine (or other sulphuric amino acids or amino acid analogous) may be added to inhibit oxidation of methionine residues to me- thionine sulfoxide when the polypeptide acting as the therapeutic agent is a polypeptide comprising at least one methionine residue susceptible to such oxidation.
  • the term "inhibit" in this context is intended to mean minimal accumulation of methionine oxidized species over time. Inhibiting methionine oxidation results in greater retention of the polypeptide in its proper molecular form. Any stereoisomer of methionine (L or D) or combinations thereof can be used.
  • the amount to be added should be an amount sufficient to inhibit oxidation of the methionine residues such that the amount of methionine sulfoxide is acceptable to regulatory agencies. Typically, this means that the composition contains no more than about 10% to about 30% methionine sulfoxide. Generally, this can be achieved by adding methionine such that the ratio of methionine added to methionine residues ranges from about 1 :1 to about 1000: 1 , such as 10: 1 to about 100: 1.
  • the formulation further or alternatively comprises a stabilizer selected from the group of high molecular weight polymers or low molecular compounds.
  • the stabilizer is selected from polyethylene glycol (e.g. PEG 3350), polyvinyl alcohol (PVA), polyvinylpyrrolidone, carboxy- /hydroxycellulose or derivates thereof (e.g. HPC, HPC-SL, HPC-L and HPMC), cyclodextrins, sulphur-containing substances as monothioglycerol, thioglycolic acid and 2- methylthioethanol, and different salts (e.g. sodium chloride).
  • PEG 3350 polyethylene glycol
  • PVA polyvinyl alcohol
  • PVpyrrolidone polyvinylpyrrolidone
  • carboxy- /hydroxycellulose or derivates thereof e.g. HPC, HPC-SL, HPC-L and HPMC
  • cyclodextrins e.g. HPC, HPC-SL, HPC-
  • compositions may also or alternatively comprise additional sta- bilizing agents, which further enhance stability of a therapeutically active polypeptide therein.
  • Stabilizing agents of particular interest to the present invention include, but are not limited to, methionine and EDTA, which protect the polypeptide against methionine oxidation, and a nonionic surfactant, which protects the polypeptide against aggregation associated with freeze-thawing or mechanical shearing.
  • the formulation further or alternatively comprises a surfactant.
  • the surfactant may, for example, be selected from a detergent, ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglycerides, sorbitan fatty acid esters, polyoxypro- pylene-polyoxyethylene block polymers (eg. poloxamers such as Pluronic ® F68, poloxamer 188 and 407, Triton X-100 ), polyoxyethylene sorbitan fatty acid esters, polyoxyethylene and polyethylene derivatives such as alkylated and alkoxylated derivatives (tweens, e.g.
  • Tween- 20, Tween-40, Tween-80 and Brij-35 monoglycerides or ethoxylated derivatives thereof, diglycerides or polyoxyethylene derivatives thereof, alcohols, glycerol, lectins and phospholipids (eg. phosphatidyl serine, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl inositol, diphosphatidyl glycerol and sphingomyelin), derivates of phospholipids (eg. dipalmi- toyl phosphatidic acid) and lysophospholipids (eg.
  • phospholipids eg. dipalmi- toyl phosphatidic acid
  • lysophospholipids eg.
  • galactopyransoide sphingoglycol- ipids (eg. ceramides, gangliosides), dodecylphosphocholine, hen egg lysolecithin, fusidic acid derivatives- (e.g.
  • fatty acids and salts thereof C6-C12 (e.g., oleic acid and caprylic acid), acylcarnitines and derivatives, NT- acylated derivatives of lysine, arginine or histidine, or side-chain acylated derivatives of lysine or arginine, NT-acylated derivatives of dipeptides comprising any combination of lysine, arginine or histidine and a neutral or acidic amino acid, N ⁇ -acylated derivative of a tripeptide comprising any combination of a neutral amino acid and two charged amino acids, DSS (do- cusate sodium, CAS registry no [577-11-7]), docusate calcium, CAS registry no [128-49-4]), docusate potassium, CAS registry no [7491 -09-0]), SDS (sodium dodecyl sulphate or sodium lauryl
  • N-alkyl-N,N-dimethylammonio-1 -propanesulfonates 3-cholamido-1- propyldimethylammonio-1 -propanesulfonate
  • cationic surfactants quaternary ammonium bases
  • cetyl-trimethylammonium bromide cetylpyridinium chloride
  • non-ionic surfactants eg. Dodecyl ⁇ -D-glucopyranoside
  • poloxamines eg.
  • Tetronic's which are tetrafunc- tional block copolymers derived from sequential addition of propylene oxide and ethylene oxide to ethylenediamine, or the surfactant may be selected from the group of imidazoline derivatives, or mixtures thereof. Each one of these specific surfactants constitutes an alternative embodiment of the invention.
  • the formulation further or alternatively comprises protease inhibitors such as EDTA (ethylenediamine tetraacetic acid) and benzamidineHCI, but other commercially available protease inhibitors may also be used.
  • protease inhibitors such as EDTA (ethylenediamine tetraacetic acid) and benzamidineHCI, but other commercially available protease inhibitors may also be used.
  • EDTA ethylenediamine tetraacetic acid
  • benzamidineHCI benzamidineHCI
  • the use of a protease inhibitor is particular useful in pharmaceutical compositions comprising zymogens of proteases in order to inhibit autocatalysis.
  • ingredients may also or alternatively be present in the peptide pharmaceutical formulation of the present invention.
  • additional ingredients may in- elude wetting agents, emulsifiers, antioxidants, bulking agents, tonicity modifiers, chelating agents, metal ions, oleaginous vehicles, proteins (e.g., human serum albumin, gelatine or proteins) and a zwitterion (e.g., an amino acid such as betaine, taurine, arginine, glycine, lysine and histidine).
  • a zwitterion e.g., an amino acid such as betaine, taurine, arginine, glycine, lysine and histidine.
  • compositions containing an antibody according to the present invention may be administered to a patient in need of such treatment at several sites, for example, at topical sites, for example, skin and mucosal sites, at sites which bypass absorption, for example, administration in an artery, in a vein, in the heart, and at sites which involve absorption, for example, administration in the skin, under the skin, in a muscle or in the abdo- men.
  • topical sites for example, skin and mucosal sites
  • sites which bypass absorption for example, administration in an artery, in a vein, in the heart
  • sites which involve absorption for example, administration in the skin, under the skin, in a muscle or in the abdo- men.
  • Administration of pharmaceutical compositions according to the invention may be through several routes of administration, for example, lingual, sublingual, buccal, in the mouth, oral, in the stomach and intestine, nasal, pulmonary, for example, through the bronchioles and alveoli or a combination thereof, epidermal, dermal, transdermal, vaginal, rectal, ocular, for examples through the conjunctiva, uretal, and parenteral to patients in need of such a treatment.
  • routes of administration for example, lingual, sublingual, buccal, in the mouth, oral, in the stomach and intestine, nasal, pulmonary, for example, through the bronchioles and alveoli or a combination thereof, epidermal, dermal, transdermal, vaginal, rectal, ocular, for examples through the conjunctiva, uretal, and parenteral to patients in need of such a treatment.
  • compositions of the current invention may be administered in several dosage forms, for example, as solutions, suspensions, emulsions, microemulsions, multiple emulsion, foams, salves, pastes, plasters, ointments, tablets, coated tablets, rinses, capsules, for ex- ample, hard gelatine capsules and soft gelatine capsules, suppositories, rectal capsules, drops, gels, sprays, powder, aerosols, inhalants, eye drops, ophthalmic ointments, ophthalmic rinses, vaginal pessaries, vaginal rings, vaginal ointments, injection solution, in situ transforming solutions, for example in situ gelling, in situ setting, in situ precipitating, in situ crystallization, infusion solution, and implants.
  • solutions for example, suspensions, emulsions, microemulsions, multiple emulsion, foams, salves, pastes, plasters, ointments, tablets, coated tablets, rinse
  • compositions of the invention may further be compounded in, or attached to, for example through covalent, hydrophobic and electrostatic interactions, a drug carrier, drug delivery system and advanced drug delivery system in order to further enhance stability of the antibody, increase bioavailability, increase solubility, decrease adverse effects, achieve chronotherapy well known to those skilled in the art, and increase patient compliance or any combination thereof.
  • Examples of carriers, drug delivery systems and advanced drug deliv- ery systems include, but are not limited to, polymers, for example cellulose and derivatives, polysaccharides, for example dextran and derivatives, starch and derivatives, polyvinyl alcohol), acrylate and methacrylate polymers, polylactic and polyglycolic acid and block copolymers thereof, polyethylene glycols, carrier proteins, for example albumin, gels, for exam- pie, thermogelling systems, for example block co-polymeric systems well known to those skilled in the art, micelles, liposomes, microspheres, nanoparticulates, liquid crystals and dispersions thereof, L2 phase and dispersions there of, well known to those skilled in the art of phase behaviour in lipid-water systems, polymeric micelles, multiple emulsions, self- emulsifying, self-microemulsifying, cyclodextrins and derivatives thereof, and dendrimers.
  • polymers for example cellulose and derivatives, polysaccharides,
  • compositions of the current invention are useful in the formulation of solids, semisolids, powder and solutions for pulmonary administration of an antibody, using, for example a metered dose inhaler, dry powder inhaler and a nebulizer, all being devices well known to those skilled in the art.
  • compositions of the current invention are specifically useful in the formulation of controlled, sustained, protracting, retarded, and slow release drug delivery systems. More specifically, but not limited to, compositions are useful in formulation of parenteral controlled release and sustained release systems (both systems leading to a many-fold reduction in number of administrations), well known to those skilled in the art. Even more preferably, are controlled release and sustained release systems administered subcutaneous.
  • parenteral controlled release and sustained release systems both systems leading to a many-fold reduction in number of administrations
  • examples of useful controlled release system and compositions are hydrogels, oleaginous gels, liquid crystals, polymeric micelles, microspheres, nanoparticles,
  • Methods to produce controlled release systems useful for compositions of the current invention include, but are not limited to, crystallization, condensation, co-crystallization, precipitation, co-precipitation, emulsification, dispersion, high pressure homogenisation, encapsulation, spray drying, microencapsulating, coacervation, phase separation, solvent evaporation to produce microspheres, extrusion and supercritical fluid processes.
  • General reference is made to Handbook of Pharmaceutical Controlled Release (Wise, D. L., ed. Marcel Dekker, New York, 2000) and Drug and the Pharmaceutical Sciences vol. 99: Protein Formulation and Delivery (MacNally, E. J., ed. Marcel Dekker, New York, 2000).
  • Parenteral administration may be performed by subcutaneous, intramuscular, intraperitoneal or intravenous injection by means of a syringe, optionally a pen-like syringe.
  • parenteral administration can be performed by means of an infusion pump.
  • a further option is a composition which may be a solution or suspension for the administration of the antibody compound in the form of a nasal or pulmonal spray.
  • the pharmaceutical compositions containing an antibody of the invention can also be adapted to transdermal administration, e.g. by needle-free injection or from a patch, optionally an ion- tophoretic patch, or transmucosal, e.g. buccal, administration.
  • the antibody can be administered via the pulmonary route in a vehicle, as a solu- tion, suspension or dry powder using any of known types of devices suitable for pulmonary drug delivery.
  • examples of these comprise of, but are not limited to, the three general types of aerosol-generating for pulmonary drug delivery, and may include jet or ultrasonic nebulizers, metered-dose inhalers, or dry powder inhalers (Cf. Yu J, Chien YW. Pulmonary drug delivery: Physiologic and mechanistic aspects. Crit Rev Ther Drug Carr Sys 14(4) (1997) 395- 453).
  • the aerodynamic diameter (d a ) of a particle is defined as the geometric equivalent diameter of a reference standard spherical particle of unit density (1 g/cm 3 ).
  • d a is related to a reference diameter (d) as a function of the square root of the density ratio as described by:
  • Mass median aerodynamic diameter (MMAD) and mass median effective aerodynamic diameter (MMEAD) are used inter-changeably, are statistical parameters, and empirically describe the size of aerosol particles in relation to their potential to deposit in the lungs, independent of actual shape, size, or density (cf. Edwards DA, Ben-Jebria A, Langer R. Recent advances in pulmonary drug delivery using large, porous inhaled particles. J Appl Physiol 84(2) (1998) 379-385).
  • MMAD is normally calculated from the measurement made with impactors, an instrument that measures the particle inertial behaviour in air.
  • the formulation could be aerosolized by any known aero- solisation technology, such as nebulisation, to achieve a MMAD of aerosol particles less than 10 ⁇ m, more preferably between 1 -5 ⁇ m, and most preferably between 1 -3 ⁇ m.
  • the preferred particle size is based on the most effective size for delivery of drug to the deep lung, where protein is optimally absorbed (cf. Edwards DA, Ben-Jebria A, Langer A, Recent advances in pulmonary drug delivery using large, porous inhaled particles. J Appl Physiol 84(2) (1998) 379-385).
  • Deep lung deposition of the pulmonal formulations comprising the antibody may op- tional be further optimized by using modifications of the inhalation techniques, for example, but not limited to: slow inhalation flow (eg. 30 L/min), breath holding and timing of actuation.
  • stabilized formulation refers to a formulation with increased physical stability, increased chemical stability or increased physical and chemical stability.
  • physical stability of the protein formulation as used herein refers to the tendency of the antibody to form biologically inactive and/or insoluble aggregates as a result of exposure of the antibody to thermo-mechanical stresses and/or interaction with interfaces and surfaces that are destabilizing, such as hydrophobic surfaces and interfaces.
  • Physical stability of the aqueous antibody formulations is evaluated by means of visual inspection and/or turbidity measurements after exposing the formulation filled in suitable containers (e.g. cartridges or vials) to mechanical/physical stress (e.g. agitation) at different temperatures for various time periods. Visual inspection of the formulations is performed in a sharp focused light with a dark background.
  • the turbidity of the formulation is characterized by a visual score ranking the degree of turbidity for instance on a scale from 0 to 3 (a formulation showing no turbidity corresponds to a visual score 0, and a formulation showing visual turbid- ity in daylight corresponds to visual score 3).
  • a formulation is classified physical unstable with respect to antibody aggregation, when it shows visual turbidity in daylight.
  • the turbidity of the formulation can be evaluated by simple turbidity measurements well- known to the skilled person. Physical stability of the aqueous antibody formulations can also be evaluated by using a spectroscopic agent or probe of the conformational status of the an- tibody.
  • the probe is preferably a small molecule that preferentially binds to a non-native con- former of the antibody.
  • a small molecular spectroscopic probe of protein structure is Thioflavin T.
  • Thioflavin T is a fluorescent dye that has been widely used for the detection of amyloid fibrils. In the presence of fibrils, and perhaps other protein configurations as well, Thioflavin T gives rise to a new excitation maximum at about 450 nm and enhanced emission at about 482 nm when bound to a fibril protein form. Unbound Thioflavin T is essentially non-fluorescent at the wavelengths.
  • hydrophobic patch probes that bind preferentially to exposed hydrophobic patches of a protein.
  • the hydrophobic patches are gener- ally buried within the tertiary structure of a protein in its native state, but become exposed as a protein begins to unfold or denature.
  • these small molecular, spectroscopic probes are aromatic, hydrophobic dyes, such as antrhacene, acridine, phenanthroline or the like.
  • spectroscopic probes are metal-amino acid complexes, such as cobalt metal complexes of hydrophobic amino acids, such as phenylalanine, leucine, isoleucine, methion- ine, and valine, or the like.
  • chemical stability of the antibody formulation refers to chemical covalent changes in the antibody structure leading to formation of chemical degradation products with potential less biological potency and/or potential increased immunogenic properties compared to the native antibody structure.
  • chemical degradation products can be formed depending on the type and nature of the native antibody and the environment to which the antibody is exposed. Elimination of chemical degradation can most probably not be completely avoided and increasing amounts of chemical degradation products is often seen during storage and use of the antibody formulation as well-known by the person skilled in the art.
  • Most proteins are prone to deamidation, a process in which the side chain amide group in glutaminyl or asparaginyl residues is hydrolysed to form a free carbox- ylic acid.
  • a “stabilized formulation” refers to a formulation with increased physical stability, increased chemical stability or increased physical and chemical stability.
  • a formulation must be stable during use and storage (in compliance with recommended use and storage conditions) until the expiration date is reached.
  • the pharmaceutical formulation comprising the antibody is stable for more than 6 weeks of usage and for more than 3 years of storage.
  • the pharmaceutical formulation comprising the antibody is stable for more than 4 weeks of usage and for more than 3 years of storage. In a further embodiment of the invention the pharmaceutical formulation comprising the antibody is stable for more than 4 weeks of usage and for more than two years of storage.
  • the pharmaceutical formulation com- prising the antibody is stable for more than 2 weeks of usage and for more than two years of storage.
  • Suitable antibody formulations can also be determined by examining experiences with other already developed therapeutic monoclonal antibodies.
  • monoclonal antibodies have been shown to be efficient in clinical situations, such as Rituxan (Rituximab), Herceptin (Trastuzumab) Xolair (Omalizumab), Bexxar (Tositumomab), Campath (Alemtuzumab), Zevalin, Oncolym, Humira and similar formulations may be used with the antibodies of this invention.
  • a monoclonal antibody can be supplied at a concentration of 10 mg/ml_ in either 100 mg (10 ml.) or 500 mg (50 ml.) single-use vials, formulated for IV administration in 9.0 mg/ml_ sodium chloride, 7.35 mg/ml_ sodium citrate dihydrate, 0.7 mg/ml_ polysorbate 80, and sterile water for injection. The pH is adjusted to 6.5.
  • the antibody can be formulated in a solution comprising histidin, sucrose, and Polysorbate 80.
  • anti-hNKG2D antibodies may also be useful in diagnostic assays for NKG2D protein, e.g. detecting its expression in specific cells, tissues, or serum.
  • anti- hNKG2D antibodies could be used in assays selecting patients for anti-hNKG2D treatment.
  • the anti-hNKG2D antibodies could be used for analyzing for the presence of hNKG2D in serum or tissue specimens, testing for the presence of CD4+ T cells ex- pressing NKG2D, or the presence of disease promoting cells expressing NKG2D (e.g., NK or CD4+ or CD8+ T cells).
  • analyses could be combined with analyses testing, e.g., for the levels of soluble MICA in blood (see, e.g., WO2003089616 by Spies et a/.).
  • the antibody typically will be labeled with a detectable moiety.
  • a detectable moiety Numerous labels are available that can be generally grouped into the following cate- gories:
  • Radioisotopes such as 35 S, 14 C, 125 I, 3 H, and 131 I.
  • the antibody can be labeled with the radioisotope using the techniques described in Current Protocols in Immunology, Volumes 1 and 2, Coligen et al. , Ed. Wiley-lnterscience, New York, N. Y., Pubs. (1991), for example, and radioactivity can be measured using scintillation counting.
  • Fluorescent labels such as rare-earth chelates (europium chelates) or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, Lissamine, phycoerythrin and Texas Red are available. The fluorescent labels can be conjugated to the antibody using the techniques disclosed in Current Protocols in Immunology, supra, for example. Fluores- cence can be quantified using a fluorimeter.
  • the enzyme generally catalyzes a chemical alteration of the chromogenic substrate that can be measured using various techniques. For example, the enzyme may catalyze a color change in a substrate, which can be measured spectropho- tometrically. Alternatively, the enzyme may alter the fluorescence or chemiluminescence of the substrate. Techniques for quantifying a change in fluorescence are described above.
  • the chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light that can be measured (using a chemiluminometer, for example) or donates energy to a fluorescent acceptor.
  • enzymatic labels include luciferases (e.g., fire- fly luciferase and bacterial luciferase; U.S. Pat. No. 4,737,456), luciferin, 2,3- dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, beta-galactosidase, glucoamylase, lysozyme, saccharide oxidases (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxi- dase, microperoxidase, and the like.
  • luciferases e.g., fire- fly luciferase and bacterial luciferase
  • enzyme-substrate combinations include, for example: (i) Horseradish peroxidase (HRPO) with hydrogen peroxidase as a substrate, wherein the hydrogen peroxidase oxidizes a dye precursor (e.g., orthophenylene diamine (OPD) or 3,3',5,5'-tetramethyl benzidine hydrochloride (TMB));
  • HRPO Horseradish peroxidase
  • OPD orthophenylene diamine
  • TMB 3,3',5,5'-tetramethyl benzidine hydrochloride
  • alkaline phosphatase AP
  • para-nitrophenyl phosphate as chromogenic substrate
  • beta-D-galactosidase beta-D-Gal
  • a chromogenic substrate e.g., p- nitrophenyl-beta-D-galactosidase
  • fluorogenic substrate 4-methylumbelliferyl-p-beta- galactosidase
  • the label is indirectly conjugated with the antibody.
  • the antibody can be conjugated with biotin, and any of the three broad categories of labels mentioned above can be conjugated with avidin, or vice versa. Biotin binds selectively to avidin, and thus, the label can be conjugated with the antibody in this indirect manner.
  • the antibody is conjugated with a small hapten (e.g., digoxin) and one of the different types of labels mentioned above is conjugated with an anti-hapten antibody (e.g., anti-digoxin antibody).
  • a small hapten e.g., digoxin
  • an anti-hapten antibody e.g., anti-digoxin antibody
  • the anti-NKG2D antibody need not be labeled, and the presence thereof can be detected using a labeled secondary antibody that binds to the NKG2D antibody.
  • the antibodies of the present invention may be employed in any known assay method, such as competitive-binding assays, direct and indirect sandwich assays, and im- munoprecipitation assays. Zola, Monoclonal Antibodies: A Manual of Techniques, pp. 147- 158 (CRC Press, Inc. 1987).
  • the tissue sample may be fresh or frozen or may be embedded in paraffin and fixed with a preservative such as formalin, for example.
  • the antibodies may also be used for in vivo diagnostic assays.
  • the anti- body is labeled with a radionuclide or a non-radioactive indicator detectable by, e.g., nuclear magnetic resonance, or other means known in the art.
  • the label is a radiolabel, such as, e.g., 125 I , 131 1, 67 Cu, 99m Tc, or 111 In.
  • the labeled antibody is administered to a host, preferably via the bloodstream, and the presence and location of the labeled antibody in the host is assayed. This imaging technique is suitably used in the detection, staging and treat- ment of neoplasms.
  • the radioisotope is conjugated to the protein by any means, including metal-chelating compounds or lactoperoxidase, or iodogen techniques for iodination.
  • the antibodies of the present invention can be provided in a kit, i.e. , a packaged combination of reagents in predetermined amounts with instructions for performing the diagnostic assay.
  • the kit will include substrates and cofactors required by the enzyme (e.g., a substrate precursor that provides the detectable chromophore or fluorophore).
  • substrates and cofactors required by the enzyme e.g., a substrate precursor that provides the detectable chromophore or fluorophore
  • other additives may be included such as stabilizers, buffers (e.g., a block buffer or lysis buffer) and the like.
  • the relative amounts of the various reagents may be varied widely to provide for concentrations in solution of the reagents that substantially optimize the sensitivity of the assay.
  • the reagents may be provided as dry powders, usually lyophilized, including excipients that on dissolution will provide a reagent solution having the appropriate concentration.
  • the invention provides for the use of a human or humanized antibody as described herein in the preparation of a pharmaceutical composition for administration to a human patient.
  • the patient suffers from, or is at risk for, an autoimmune or inflammatory disease or disorder.
  • the invention provides a method of reducing or inhibit- ing hNKG2D-mediated activition of NK or T cells in a patient in need thereof, comprising the step of administering a human or humanized anti-NKG2D antibody to the patient, which antibody reduces or prevents ligand-mediated activation of the NKG2D receptor.
  • the method directed at decreasing the activity of such lymphocytes in patients having a disease in which increased NK or T cell activity is detrimental which involves, affects or is caused by cells susceptible to lysis by NK or T cells, or which is caused or characterized by increased NK and/or T cell activity, such as an autoimmune disease or disorder or an inflammatory condition.
  • the invention provides a method of reducing chronic inflammation in the patient.
  • Exemplary conditions or disorders to be treated with the polypeptides, antibodies and other compounds of the invention include, but are not limited to systemic lupus erythe- matosis, rheumatoid arthritis, juvenile chronic arthritis, psoriatic arthritis, osteoarthritis, spondyloarthropathies (ankylosing spondylitis), systemic sclerosis (scleroderma), idiopathic inflammatory myopathies (dermatomyositis, polymyositis), Sjogren's syndrome, vasculitis, systemic vasculitis, temporal arteritis, atherosclerosis, sarcoidosis, myasthenia gravis, auto- immune hemolytic anemia (immune pancytopenia, paroxysmal nocturnal hemoglobinuria), pernicious anemia, autoimmune thrombocytopenia (idiopathic thrombocytopenic purpura, immune-mediated thrombocytopenia), thyroiditis (Grave's disease,
  • autoimmune chronic active hepatitis viral hepatitis, pri- mary biliary cirrhosis, granulomatous hepatitis, Wegener's granulomatosis, Behcet's disease, and sclerosing cholangitis
  • inflammatory bowel diseases such as ulcerative colitis or Crohn's disease, celiac disease, gluten-sensitive enteropathy, and Whipple's disease
  • autoimmune or immune-mediated skin diseases including bullous skin diseases, erythema multiforme and contact dermatitis, dermitis herpetiformis, psoriasis, pemphigus vulgaris, vitiligo (leukoderma), allergic diseases such as asthma, allergic rhinitis, atopic dermatitis, food hypersensitivity and urticaria, immunologic diseases of the lung such as eosinophilic pneumonias, idiopathic pulmonary fibrosis and hypersensitivity pneumoni
  • the anti-NKG2D antibody is used in combination with one or more other anti-inflammatory agents, including, but not limited to, analgesic agents, immunosuppressive agents (e.g., B- or T-cell antagonists such as B-cell depletion agents and T cell inhibiting agents; complement inhibiting agents), corticosteroids, and anti- TNFalpha agents or other anti-cytokine or anti-cytokine receptor agents, and anti-angiogenic agents.
  • analgesic agents e.g., immunosuppressive agents (e.g., B- or T-cell antagonists such as B-cell depletion agents and T cell inhibiting agents; complement inhibiting agents), corticosteroids, and anti- TNFalpha agents or other anti-cytokine or anti-cytokine receptor agents, and anti-angiogenic agents.
  • immunosuppressive agents e.g., B- or T-cell antagonists such as B-cell depletion agents and T cell inhibiting agents; complement inhibiting agents
  • corticosteroids e
  • the NKG2D-based treatment may precede, or follow, the other treatment by, e.g., intervals ranging from minutes to weeks and months. It also is envisioned that more than one administration of either the anti-NKG2D composition or the other agent will be utilized.
  • the agents may be administered interchangeably, on alternate days or weeks; or a cycle of anti- NKG2D treatment may be given, followed by a cycle of the other agent therapy. In any event, all that is required is to deliver both agents in a combined amount effective to exert a therapeutically beneficial effect, irrespective of the times for administration.
  • the following describes some selected inflammatory and/or autoimmune diseases or disorders for which anti-hNKG2D antibodies of the invention can be used as therapeutic agents.
  • the anti-hNKG2D antibody is full-length bivalent MS or 21 F2, or an antigen-binding fragment, variant or derivative thereof.
  • Rheumatoid arthritis is a chronic systemic autoimmune inflammatory disease that mainly involves the synovial membrane of multiple joints with resultant injury to the articular cartilage.
  • the pathogenesis is T lymphocyte dependent and is associated with the production of rheumatoid factors, auto antibodies directed against self IgG, with the resultant formation of immune complexes that attain high levels in joint fluid and blood. These complexes in the joint may induce the marked infiltrate of lymphocytes and monocytes into the synovium and subsequent marked synovial changes; the joint space is infiltrated by similar cells with the addition of numerous neutrophils.
  • the pathological T cell produces cytokines and other soluble factors adding to the attraction and activation of other cells, and to the destruction of the tissue.
  • Tissues affected are primarily the joints, often in symmetrical pattern.
  • extra-articular disease also occurs in two major forms.
  • One form is the develop- ment of extra-articular lesions with ongoing progressive joint disease and typical lesions of pulmonary fibrosis, vasculitis, and cutaneous ulcers.
  • the second form of extra-articular disease is the so called Felty's syndrome which occurs late in the RA disease course, sometimes after joint disease has become quiescent, and involves the presence of neutropenia, thrombocytopenia and splenomegaly. This can be accompanied by vasculitis in multiple or- gans with formations of infarcts, skin ulcers and gangrene.
  • the invention provides a method for treating and/or preventing rheumatoid arthritis (RA).
  • the method comprises delivering an effective amount of an anti-hNKG2D antibody to a patient having RA or being identified/diagnosed as being at substantial risk of developing RA, such that RA is treated or prevented.
  • the anti- NKG2D antibody is demonstrated to be effective in ameliorating RA in an acceptable model of RA, such as is described in US Patent No. 6414218 and US Patent Publication No.
  • the antibody that is capable of detectably reducing ligand-induced NKG2D activation of NKG2D-expressing leukocytes and/or impairing expansion of NKG2D+ T cells or NK cells (e.g., impairing the expansion and/or function of autoreactive CD8+ T cells) (in contrast to, e.g., at least some of the antibodies described in US Patent Publication No. 20040115198), without significantly depleting such cells (e;g., causing a reduction of about 10% or less of such cells as compared to a suitable control).
  • the method results in a modulation of one or more biomarkers in a manner consistent with the treatment or prevention (as applicable) of RA (e.g., serum IL-6, TNF- ⁇ , IL-1 , VEGF, TIFF R, IL-2R, shed CD4, shed CD8, and/or C reactive protein).
  • RA e.g., serum IL-6, TNF- ⁇ , IL-1 , VEGF, TIFF R, IL-2R, shed CD4, shed CD8, and/or C reactive protein.
  • the prac- tice of the method results in a detectable reduction of synovial inflammation in the peripheral joints of the patient/host.
  • the method results in preventing radiographic deterioration and improving physical function in the patient or host as exhibited by, e.g., a reduction in radiographic progression in the patient or host, reduction in swollen and tender joints (as determined by acceptable analytical criteria), and/or significantly improved quality of life (e.g., as determined by a reduction in disability scores on the RA Health Assessment Questionnaire).
  • the antibody can be used alone or in combination with one or more other anti-RA agent, such as a non-steroidal anti-inflammatory drug (NSAID), a COX-2 inhibitor, an analgesic, a corticosteroid (e.g., predinisone, hydrocortisone), gold, an immunosuppressant (e.g., methotrexate), a B-cell depletion agent (e.g., Rituxan®), a B-cell agonist (e.g., LymphoStat- B®) and an anti-TNFalpha agent (e.g., Embrel®, Humira® and Remicade®), an anti-IL1 receptor antagonist (e.g., Kineret®), an anti-IL-15 antibody, or a disease-modifying antirheumatic drug (DMARD).
  • NSAID non-steroidal anti-inflammatory drug
  • COX-2 inhibitor e.g., a COX-2 inhibitor
  • an analgesic e.g., analges
  • Demyelinating diseases of the central and peripheral nervous systems including Multiple Sclerosis (MS); idiopathic demyelinating polyneuropathy or Guillain-Barre syndrome; and Chronic Inflammatory Demyelinating Polyneuropathy, are believed to have an autoimmune basis and result in nerve demyelination as a result of damage caused to oligodendrocytes or to myelin directly.
  • MS is a demyelinating disease that is T lym- phocyte-dependent and has either a relapsing-remitting course or a chronic progressive course.
  • the invention provides a method for treating and/or preventing MS.
  • the method comprises delivering an effective amount of an anti-hNKG2D antibody to a human patient having MS or being identified/diagnosed as being at substantial risk of developing MS, such that MS is treated or prevented in the patient or host.
  • the anti-NKG2D monoclonal antibody is capable of detectably reducing ligand-induced NKG2D activation of NKG2D-expression leukocytes and/or impairing expansion of NKG2D+ T cells or NK cells, without significantly depleting such cells.
  • the antibody can be used alone or in combination with other anti-MS agents such as Tyzabri®. Inflammatory bowel disease In CD8 + T cells in the intestine, NKG2D acts as a co-stimulator of CD28 " cells (Roberts et al., J Immunol 2001 ; 167:5527-30).
  • NKG2D is upregulated, and intestinal epithelial lymphocytes are stimulated via NKG2D to kill and produce cytokines (Hue et al., Immunity 2004;21 :367-77; Meresse et al., Immunity 2004;21 :357-66). Additionally, IL-15, often found during intestinal inflammation, upregulates NKG2D on intestinal epithelial lymphocytes (Roberts et al., J Immunology
  • the invention provides a method for treating and/or preventing inflammatory bowel disease (IBD), such as Crohn's disease or ulcerative colitis.
  • IBD inflammatory bowel disease
  • the method of treating an inflammatory bowel disease comprises delivering an effective amount of an anti-NKG2D antibody to a human patient having IBD or being identified/diagnosed as being at substantial risk of developing IBD, such that IBD is treated or prevented in the patient.
  • the inventive IBD treatment/prevention method is practiced by use of an anti-NKG2D monoclonal antibody that is capable of detectably reduc- ing ligand-induced NKG2D activation of NKG2D-expressing leukocytes and/or impairing expansion of NKG2D+ T cells or NK cells, without significantly depleting such cells.
  • the antibody can be used alone or in combination with other anti-IBD agents, such as drugs containing mesalamine (including sulfasalazine and other agents containing 5-aminosalicylic acid (5- ASA), such as olsalazine and balsalazide), non-steroidal anti-inflammatory drugs (NSAIDs), analgesics, corticosteroids (e.g., predinisone, hydrocortisone), TNF-inhibitors (including adilimumab (Humira®, etanercept (Enbrel® and infliximab (Remicade®), anti-IL12 antibodies, immunosuppressants (such as 6-mercaptopurine, azathioprine and cyclosporine A), and antibiotics.
  • drugs containing mesalamine including sulfasalazine and other agents containing 5-aminosalicylic acid (5- ASA), such as olsalazine and balsalazide
  • Psoriasis is a T lymphocyte-mediated inflammatory disease. Lesions contain infiltrates of T lymphocytes, macrophages and antigen processing cells, and some neutrophils.
  • the invention provides a method for treating and/or preventing psoriasis.
  • the method comprises delivering an effective amount of an anti-hNKG2D antibody to a human patient having psoriasis or being identified/diagnosed as being at substan- tial risk of developing psoriasis, such that psoriasis is treated or prevented in the patient.
  • the agent is an anti-NKG2D monoclonal antibody that is capable of detectably reducing ligand-induced NKG2D activation of NKG2D-expressing leukocytes and/or impairing expansion of NKG2D+ T cells or NK cells, without significantly depleting such cells.
  • the antibody can be used alone or in combination with one or more other anti- psoriasis treatments such as phototherapy, topical therapy (e.g., tar, topical glucocorticoids), or systemic therapy (e.g., methotrexate, a synthetic retinoid, cyclosporine), an anti-TNFalpha agent (e.g. , Embrel®, Humira®, Remicade®), a T-cell inhibitor (e.g., Raptiva®), vitamin D analogs, p38 mitogen-activated protein kinase (MAPK) inhibitors, as well as a biologic agent such as Rituxan®.
  • topical therapy e.g., tar, topical glucocorticoids
  • systemic therapy e.g., methotrexate, a synthetic retinoid, cyclosporine
  • an anti-TNFalpha agent e.g. , Embrel®, Humira®, Remicade®
  • Psoriatic arthritis is a chronic inflammatory arthritic condition affecting the skin, the joints, the insertion sites of tendons, ligaments, and fascia, and is commonly associated with psoriasis, (approximately 7% of patients with psoriasis develop psoriatic arthritis).
  • psoriatic arthritis Much evidence suggests that a T-cell-mediated process drives the pathophysiology of psoriatic arthri- tis.
  • Monocytes also play a role in psoriatic arthritis and are responsible for the production of matrix metalloproteinases, which may mediate the destructive changes in the joints of patients with psoriatic arthritis.
  • NK cells are also found in affected joints, suggesting a role in the disease pathology.
  • the invention provides a method for treating and/or prevent- ing psoriatic arthritis.
  • the method comprises delivering an effective amount of an anti- hNKG2D antibody to a human patient having psoriatic arthritis or being identified/diagnosed as being at substantial risk of developing psoriatic arthritis, such that the psoriatic arthritis is treated or prevented in the patient.
  • the agent is an anti-NKG2D monoclonal antibody that is capable of detectably reducing ligand-induced NKG2D activation of NKG2D-expressing leukocytes and/or impairing expansion of NKG2D+ T cells or NK cells, without significantly depleting such cells.
  • the antibody can be used alone or in combination with one or more other anti-psoriatic arthritis treatments such as nonsteroidal antiinflammatory drugs (aspirin, ibuprofen), methotrexate, a synthetic retinoid, cyclosporine, a corticosteroid, an anti-TNFalpha agent (e.g., Embrel®, Humira®, Remicade®).
  • nonsteroidal antiinflammatory drugs aspirin, ibuprofen
  • methotrexate e.g., methotrexate
  • a synthetic retinoid e.g., cyclosporine
  • corticosteroid e.g., a corticosteroid
  • an anti-TNFalpha agent e.g., Embrel®, Humira®, Remicade®.
  • T lymphocytes In systemic lupus erythematosus (SLE), the central mediator of disease is the production of auto-reactive antibodies to self proteins/tissues and the subsequent generation of immune-mediated inflammation. Antibodies either directly or indirectly mediate tissue injury. Though T lymphocytes have not been shown to be directly involved in tissue damage, T lymphocytes are required for the development of auto-reactive antibodies. The genesis of the disease is thus T lymphocyte dependent. Multiple organs and systems are affected clinically including kidney, lung, musculoskeletal system, mucocutaneous, eye, central nervous system, cardiovascular system, gastrointestinal tract, bone marrow and blood.
  • the invention provides a method for treating and/or prevent- ing SLE.
  • the method comprises delivering an effective amount of an anti-hNKG2D antibody to a human patient having SLE or being identified/diagnosed as being at substantial risk of developing SLE, such that the SLE is treated or prevented in the patient.
  • the agent is an anti-NKG2D monoclonal antibody that is capable of detectably reducing ligand-induced NKG2D activation of NKG2D-expressing leukocytes and/or impairing ex- pansion of NKG2D+ T cells or NK cells, without significantly depleting such cells.
  • the antibody can be used alone or in combination with other anti-SLE agents, such as non-steroidal anti-inflammatory drugs (NSAIDs), analgesics, corticosteroids (e.g., predinisone, hydrocortisone), immunosuppressants (such as cyclophosphamide, azathioprine, and methotrexate), antimalarials (such as hydroxychloroquine) and biologic drugs that inhibit the production of dsDNA antibodies (e.g. LIP 394).
  • NSAIDs non-steroidal anti-inflammatory drugs
  • analgesics e.g., analgesics
  • corticosteroids e.g., predinisone, hydrocortisone
  • immunosuppressants such as cyclophosphamide, azathioprine, and methotrexate
  • antimalarials such as hydroxychloroquine
  • biologic drugs that inhibit the production of dsDNA antibodies
  • Type I diabetes mellitus or insulin-dependent diabetes is the autoimmune destruction of pancreatic islet B cells; this destruction is mediated by auto-antibodies and autoreactive T cells.
  • Antibodies to insulin or the insulin receptor can also produce the phenotype of insulin-non-responsiveness.
  • an anti-NKG2D antibody is delivered to a patient suffering from or at substantial risk of developing type I diabetes mellitus in an amount and under conditions sufficient to treat or prevent the condition in the patient.
  • the antibody can be used alone or in combination with other anti-diabetic agents, such as insulin, or beta cell growth or survival factors, or immunomodulatory antibodies such as anti-CD3 antibodies.
  • Transplantation associated diseases including graft rejection and Graft-Versus- Host-Disease (GVHD) are T lymphocyte-dependent; inhibition of T lymphocyte function is ameliorative.
  • the invention provides methods of reducing the likelihood of transplant rejection (or reducing the severity or prolonging the time to onset of a transplant rejection-related condition, i.e., to prolong allograft survival).
  • the method comprises delivering an effective amount of an anti-hNKG2D antibody to a human patient that is about to be, is, or recently was the recipient of a tissue/organ transplant, such that the likelihood of rejec- tion is detectably reduced (e.g., as compared to a control).
  • the anti- NKG2D antibody is capable of detectably reducing ligand-induced NKG2D activation of NKG2D-expression leukocytes and/or impairing expansion of NKG2D+ T cells or NK cells, without significantly depleting such cells.
  • tissue transplants that can be treated include, but are not limited to, liver, lung, kidney, heart, small bowel, and pancreatic islet cells, as well as in bone marrow-transplantation and in the treatment of graft versus host disease (GVHD).
  • the antibody can be used alone or in combination with other agents for inhibiting transplant rejection, such as immunosuppressive agents (e.g.
  • cyclosporine azathio- prine, methylprednisolone, prednisolone, prednisone, mycophenolate mofetil, sirilimus, ra- pamycin, tacrolimus
  • anti-infective agents e.g., acyclovir, clotrimazole, ganciclovir, nystatin, trimethoprimsulfarnethoxazole
  • diuretics e.g. bumetanide, furosemide, metolazone
  • ulcer medications e.g., cimetidine, famotidine, lansoprazole, omeprazole, ranitidine, sucralfate.
  • hematopoietic growth factor(s) e.g., erythropoietin, G-CSF, GM-CSF, IL-3, IL-11 , thrombopoietin, etc.
  • antimicrobial(s) e.g., antibiotic, antiviral, antifungal
  • Other autoimmune or inflammatory diseases e.g., erythropoietin, G-CSF, GM-CSF, IL-3, IL-11 , thrombopoietin, etc.
  • antimicrobial(s) e.g., antibiotic, antiviral, antifungal
  • the invention provides methods for treating and/or preventing other autoimmune or inflammatory diseases or disorders, comprising delivering an effective amount of an anti-hNKG2D antibody to a human patient having the disease or disorder or being identified/diagnosed as being at substantial risk of developing the disease or disorder, such that it is treated or prevented in the patient, where the disease or disorder is one described below.
  • the agent is an anti-NKG2D monoclonal antibody that is capable of detectably reducing ligand-induced NKG2D activation of NKG2D- expressing leukocytes and/or impairing expansion of NKG2D+ T cells or NK cells, without significantly depleting such cells.
  • the antibody can be used alone or in combination with one or more other therapeutic agents used for treating the disease or disorder.
  • Juvenile chronic arthritis is a chronic idiopathic inflammatory disease which begins often at less than 16 years of age. Its phenotype has some similarities to RA; some patients which are rhematoid factor positive are classified as juvenile rheumatoid arthritis. The disease is sub-classified into three major categories: pauciarticular, polyarticular, and systemic. The arthritis can be severe and is typically destructive and leads to joint ankylosis and retarded growth. Other manifestations can include chronic anterior uveitis and systemic amyloidosis.
  • Spondyloarthropathies are a group of disorders with some common clinical features and the common association with the expression of HLA-B27 gene product.
  • the disorders include: ankylosing sponylitis, Reiter's syndrome (reactive arthritis), arthritis associated with inflammatory bowel disease, spondylitis associated with psoriasis, juvenile onset spondyloarthropathy and undifferentiated spondyloarthropathy.
  • Distinguishing features include sacroileitis with or without spondylitis; inflammatory asymmetric arthritis; association with HLA- B27 (a serologically defined allele of the HLA-B locus of class I MHC); ocular inflammation, and absence of autoantibodies associated with other rheumatoid disease.
  • the cell most implicated as key to induction of the disease is the CD8+ T lymphocyte, a cell which targets antigen presented by class I MHC molecules.
  • CD8+ T cells may react against the class I MHC allele HLA B27 as if it were a foreign peptide expressed by MHC class I molecules. It has been hypothesized that an epitope of HLA-B27 may mimic a bacterial or other microbial antigenic epitope and thus induce a CD8+ T cells response.
  • Systemic sclerosis has an unknown etiology.
  • a hallmark of the disease is induration of the skin; likely this is induced by an active inflammatory process.
  • Scleroderma can be localized or systemic; vascular lesions are common and endothelial cell injury in the microvasculature is an early and important event in the development of systemic sclerosis; the vascular injury may be immune mediated.
  • An immunologic basis is implied by the presence of mononuclear cell infiltrates in the cutaneous lesions and the presence of anti-nuclear antibodies in many patients.
  • ICAM-1 is often unregulated on the cell surface of fibroblasts in skin lesions suggesting that T cell interaction with these cells may have a role in the pathogenesis of the disease.
  • Idiopathic inflammatory myopathies including dermatomyositis, polymyositis and others are disorders of chronic muscle inflammation of unknown etiology resulting in muscle weakness. Muscle injury/inflammation is often symmetric and progressive. Autoantibodies are associated with most forms. These myositis-specific autoantibodies are directed against and inhibit the function of components, proteins and RNA's, involved in protein synthesis.
  • Sjogren's syndrome is due to immune-mediated inflammation and subsequent functional destruction of the tear glands and salivary glands.
  • the disease can be associated with or accompanied by inflammatory connective tissue diseases.
  • the disease is associated with autoantibody production against Ro and La antigens, both of which are small RNA-protein complexes. Lesions result in keratoconjunctivitis sicca, xerostomia, with other manifestations or associations including bilary cirrhosis, peripheral or sensory neuropathy, and palpable purpura.
  • Systemic vasculitis are diseases in which the primary lesion is inflammation and subsequent damage to blood vessels which results in ischemia/necrosis/degeneration to tis- sues supplied by the affected vessels and eventual end-organ dysfunction in some cases.
  • Vasculitides can also occur as a secondary lesion or sequelae to other immune-inflammatory mediated diseases such as rheumatoid arthritis, systemic sclerosis, etc., particularly in diseases also associated with the formation of immune complexes.
  • Systemic necrotizing vasculitis polyarteritis nodosa, allergic angiitis and granulomatosis, polyangiitis; Wegener's granulomatosis; lymphomatoid granulomatosis; and giant cell arteritis.
  • Miscellaneous vasculitides include: mucocutaneous lymph node syndrome (MLNS or Kawasaki's disease), isolated CNS vasculitis, Behet's disease, thromboangiitis obliterans (Buerger's disease) and cutaneous necrotizing venulitis.
  • MLNS mucocutaneous lymph node syndrome
  • isolated CNS vasculitis Behet's disease
  • thromboangiitis obliterans Buerger's disease
  • cutaneous necrotizing venulitis The pathogenic mechanism of most of the types of vasculitis listed is believed to be primarily due to the deposition of immunoglobulin complexes in the vessel wall and subsequent induction of an inflammatory response either via
  • Sarcoidosis is a condition of unknown etiology which is characterized by the presence of epithelioid granulomas in nearly any tissue in the body; involvement of the lung is most common.
  • the pathogenesis involves the persistence of activated macrophages and lymphoid cells at sites of the disease with subsequent chronic sequelae resultant from the release of locally and systemically active products released by these cell types.
  • Autoimmune hemolytic anemia including autoimmune hemolytic anemia, immune pancytopenia, and paroxysmal noctural hemoglobinuria is a result of production of antibodies that react with antigens expressed on the surface of red blood cells (and in some cases other blood cells including platelets as well) and is a reflection of the removal of those antibody coated cells via complement mediated lysis and/or ADCC/Fc-receptor-mediated mechanisms.
  • platelet destruction/removal occurs as a result of either antibody or complement attaching to platelets and subsequent removal by complement lysis, ADCC or Fc-receptor mediated mechanisms.
  • Thyroiditis including Grave's disease, Hashimoto's thyroiditis, juvenile lymphocytic thyroiditis, and atrophic thyroiditis, are the result of an autoimmune response against thyroid antigens with production of antibodies that react with proteins present in and often specific for the thyroid gland.
  • Experimental models exist including spontaneous models: rats (BUF and BB rats) and chickens (obese chicken strain); inducible models: immunization of animals with either thyroglobulin, thyroid microsomal antigen (thyroid peroxidase).
  • Immune mediated renal diseases including glomerulonephritis and tubulointerstitial nephritis, are the result of antibody or T lymphocyte mediated injury to renal tissue either di- rectly as a result of the production of autoreactive antibodies or T cells against renal antigens or indirectly as a result of the deposition of antibodies and/or immune complexes in the kidney that are reactive against other, non-renal antigens.
  • immune-mediated diseases that result in the formation of immune-complexes can also induce immune mediated renal disease as an indirect sequelae.
  • Both direct and indirect immune mechanisms result in inflammatory response that produces/induces lesion development in renal tissues with resultant organ function impairment and in some cases progression to renal failure.
  • Both humoral and cellular immune mechanisms can be involved in the pathogenesis of lesions.
  • Inflammatory and Fibrotic Lung Disease including Eosinophilic Pneumonias; Idiopathic Pulmonary Fibrosis, and Hypersensitivity Pneumonitis may involve a disregulated im- mune-inflammatory response. Inhibition of that response would be of therapeutic benefit.
  • Autoimmune or Immune-mediated Skin Disease including Bullous Skin Diseases, Erythema Multiforme, and Contact Dermatitis are mediated by auto-antibodies, the genesis of which is T lymphocyte dependent.
  • Allergic diseases including asthma; allergic rhinitis; atopic dermatitis; food hyper- sensitivity; and urticaria are T lymphocyte dependent. These diseases are predominantly mediated by T lymphocyte induced inflammation, IgE mediated-inflammation or a combination of both.
  • Another disease suitable for treatment with a human anti-NKG2D antibody is viral hepatitis, as shown in WO2007130642 and by Chen et al. (Hepatology, Vol. 46 (3) pp. 706- 715 (2007)).
  • the effective amount of the NKG2D modulator, as well as the overall dosage regimen may vary according to the disease and the patient's clinical status, which, in turn, may be reflected in one or more clinical parameters such as clinically accepted disease scores.
  • the severity of disease and/or outcome of treatment may be evaluated by monitoring number of swollen joints; pain; mobility; and/or the official disease score ACR 20/50 or 70.
  • severity of disease and/or outcome of treatment may be evaluated by measuring blood glucose levels or variations thereof, HbIC levels, the amount of insulin needed, and the like.
  • brain inflammation can be assessed through scanning the brain.
  • severity of the disease (failure to engraft) and/or outcome of treatment may be evaluated by evidence of prolonged neutropenia, thrombocytopenia, and red-cell transfusion dependence in patients that have undergone myeloablative conditioning, and by failure to observe chimerism in patients that have undergone non-myeloablative conditioning.
  • detectable effects on treatment outcome using the methods and compositions of the present invention include a decrease in the necessity for other treatments (including, e.g., a decrease in the amount and/or duration of other drugs or treatments), a decrease in number and/or duration of hospital stays, a decrease in lost work days due to illness, and the like.
  • the effective amount may be determined by those of ordinary skill in the art by routine experimentation, by constructing a matrix of values and testing dif- ferent points in the matrix.
  • the dosage ranges from about 0.0001 to 100 mg/kg, and more usually 0.01 to 5 mg/kg, of the host body weight.
  • dosages can be about 0.3 mg/kg body weight, about 1 mg/kg body weight, about 3 mg/kg body weight, about 5 mg/kg body weight or about 10 mg/kg body weight or within the range of 1- 10 mg/kg.
  • An exemplary treatment regime entails administration twice per week, once per week, once every two weeks, once every three weeks, once every four weeks, once a month, once every 3 months or once every three to 6 months.
  • Preferred dosage regimens for an anti-hNKG2D antibody of the invention include about 1 , 3 or 10 mg/kg body weight body weight via intravenous administration or subcutaneous injection, with the antibody being given using one of the following dosing schedules: (i) loading doses every 1-3 weeks for 2-4 dosages, then every two; months (ii) every four weeks; (iii) every week, or any other optimal dosing.
  • two or more monoclonal antibodies with different binding specificities are administered simultaneously, in which case the dosage of each antibody adminis- tered falls within the ranges indicated.
  • Antibody is usually administered on multiple occa- sions. Intervals between single dosages can be, for example, weekly, monthly, every three months or yearly.
  • Intervals can also be irregular as indicated by measuring blood levels of antibody to the target antigen in the patient.
  • dosage is adjusted to achieve a plasma antibody concentration of about 1 -1000 ⁇ g /ml and in some methods about 25-300 ⁇ g/ml.
  • antibody can be administered as a sustained release formulation, in which case less frequent administration is required. Dosage and frequency vary depending on the half-life of the antibody in the patient. In general, human antibodies show the longest half life, followed by humanized antibodies, chimeric antibodies, and nonhuman antibodies. The dosage and frequency of administration can vary depending on whether the treatment is prophylactic or non-prophylactic (e.g., palliative or curative).
  • a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In palliative or curative applications, a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the pa- tient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime.
  • anti-inflammatory agents will approximate those already employed in clinical therapies wherein the anti-inflammatory agents are administered alone or in combination with other agents. Variation in dosage will likely occur depending on the condition being treated. The physician administering treatment will be able to determine the appropriate dose for the individual subject.
  • an article of manufacture containing materials useful for the treatment of the disorders described above can comprise a container containing a human or humanized anti- hNKG2D antibody as described herein together with instructions directing a user to treat a disorder such as an autoimmune or inflammatory disease or disorder in a human with the antibody in an effective amount.
  • the article of manufacture typically comprises a container and a label or package insert on or associated with the container. Suitable containers in- elude, for example, bottles, vials, syringes, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition that is effective for treating the condition and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is the human or humanized anti- hNKG2D antibody herein, or an antigen-binding fragment or antibody derivative (e.g., an im- munoconjugate) comprising such an antibody.
  • the label or package insert indicates that the composition is used for treating the condition of choice, such as, e.g., rheumatoid arthiritis.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises the human or humanized an- tibody herein, and (b) a second container with a composition contained therein, wherein the composition comprises a therapeutic agent other than the human or humanized antibody.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the first and second compositions can be used in combination to treat an autoimmune or inflammatory disease or disorder.
  • Such therapeutic agents may be any of the adjunct therapies described in the preceding section..
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as phosphate-buffered saline, Ringer's solution and dextrose solution.
  • dextrose solution such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution
  • Example 1 Generation and initial screening of human monoclonal antibodies against hNKG2D Materials and Methods
  • Soluble NKG2D-hFc fusion protein (R&D, cat: 1299-NK) or NKG2D expressed on the surface of cells (NK, BAF, or CHO) were used as antigens for immunization.
  • the BAF cells were co-transfected with full-length NKG2D and DAP10.
  • the CHO cells were transfected with an NKG2D point mutant that transports to the cell surface without DAP10 (Wu et al. , Science 1999;385:730-2).
  • the NK cells were primary NK cells naturally expressing NKG2D.
  • mice Fully human monoclonal antibodies against NKG2D were produced in the KM mouseTM strain of transgenic mice that express human antibody genes (PCT publication WO 02/43478 to lshida et al.).
  • the endogenous kappa light chain gene has been homozygously disrupted as described in Chen et al (1993) EMBO J. 12:811 -820, and the endogenous mouse heavy chain has been homozygously disrupted as described in Example 1 of PCT Publication WO 01/09187 for Humab mice.
  • the mouse strain carries a human kappa light chain transgene, KC05, as described in Fishwild et al (1996) Nature Bio- technology 14:845-851.
  • the mouse strain also carries a human heavy chain transchromo- some, SC20, as described in WO0243478.
  • mice were immunized intraperitoneal ⁇ with alternating injections of NKG2D-transfected BAF cells and NKG2D-transfected CHO cells, or primary human NK cells with or without any adjuvant. Each mouse was immunized IP with 5x10 6 cells every or every other week (6 times in total). The mice were boosted with 5x10 6 NKG2D-transfected BAF cells intravenously 3 and 2 days before sacrifice and removal of the spleen. The animal experiments were performed according to Danish National Research Council guidelines.
  • animals were immunized intraperitoneal ⁇ and in the foot path with NKG2D-hFc with different adjuvant.
  • Each mouse was immunized 7 x 25ug NKG2F-hFc/Ribi/ip/sc, 1 x 25ug NKG2D-hFc/CFA/ip/sc, 1 x 25ug NKG2D- hFc/IFA/ip/sc, 1 x 30ug anti-CTLA4 + 40ug NKG2D-hFc/IFA/ip/sc, 1 x 25ug NKG2D- hFc/Ribi/ip/sc and boosted 2 x 30ug/PBS/ip/iv 3 and 2 days before sacrifice and removal of the spleen.
  • the animal experiments were performed according to American National Research Council guidelines.
  • mice were screened by flow cytometry analysis for NKG2D-specificity and selected sera were also tested for their ability to neutralize binding of the MICA ligand, as described in Example 3. Mice that had generated high titers of antibodies that specifically bound NKG2D and neutralized MICA binding were selected for hybridoma production.
  • Hybridomas The spleen from each selected immunized mouse was homogenised and a single cell suspension of splenocytes used for fusion to X61 Ag8653 myeloma cells (ATCC, CRL 1580). The fusions were performed using polyethyleneglycol (PEG) 1500 as previously described (Harlow and Lane, ANTIBODIES: A LABORATORY
  • the fused cells were initially seeded in 96-well tissue culture plates in selective DMEM HAT medium, supplemented with 10% FBS and 5% origin (Hybridoma cloning Factor, BioVeris). The plates were incubated for 10-14 days with 1-2 medium changes, respectively, to DMEM HT medium supplemented with 5% FBS and 0.7% origin, before harvest and screening of the supernatants. Clones tested positive were expanded and subcloned by limiting dilution until stable clones had been generated. The selected clones were continuously screened for the presence of anti-NKG2D specific antibodies by FACS analysis as well as for their ability to neutralize MICA binding.
  • Hybridoma Supernatants Screening of Hybridoma Supernatants .
  • the primary screening of the hybridoma su- pernatants from the first series of immunizations was performed using direct ELISA or flow cytometry analysis (FACS) to test for the presence of anti-NKG2D specific antibodies.
  • FACS flow cytometry analysis
  • the ELISA was performed by coating maxisorp plates with 50 ⁇ l 0.4 ⁇ g/ml mFc-NKG2D (comprising the extracellular portion of NKG2D fused to murine Fc and expressed in CHO cells) overnight in PBS at 4 0 C, followed by blocking with PBS, 0.05% Tween 20, for 15 min at room temperature.
  • the plates were subsequently incubated with 50 ⁇ l hybridoma super- natant, and NKG2D-specific antibodies detected using Goat-Anti-human IgG-HRP Fey Fragment specific (Jackson, 109-036-098). These incubations were performed for 1 hr at room temperature, and between each step the plates were washed with PBS, 0.05% Tween 20. Bound antibodies were visualized using 10O ⁇ l TMB substrate (Kem-En-Tec), and stopped with 4M H 3 PO 4 . The plates were read at 450 and 620 nm.
  • binding to NKG2D-expressing BaF/3 cells and control BaF/3 cells not expressing NKG2D was analyzed by incubation of 50000 cells in 10 ⁇ l with 90 ⁇ l hybridoma supernatant for 30 min at 4 0 C, followed by washing with PBS with 2% FCS, and subsequently incubated with secondary Goat- Anti-human IgG-HRP Fey Fragment specific (Jackson, 109-036-098). The cells were then analysed on a B&D FACSArray (BD Biosciences). Antibodies that only stained NKG2D- expressing BaF/3 cells and not control cells were deemed NKG2D-specific.
  • the primary screen for the second series of immunizations was a direct ELISA to test for the presence of anti-NKG2D specific antibodies. Briefly, the ELISA was performed by coating maxisorp plates with 1-2 mg/ml hFc-NKG2D (R&D Systems) overnight in PBS at 4 0 C, followed by blocking with PBS, 0.05% Tween 20, 5% chicken serum for 30-60 min at room temperature. The plates were subsequently incubated with 50 ⁇ l hybridoma supernatant and 50 ⁇ l blocking buffer, and NKG2D-specific antibodies detected using Anti-human IgG-HRP (Bethyl, A80-1 15P) in blocoking buffer.
  • Anti-human IgG-HRP Bethyl, A80-1 15P
  • Hybridomas selected from an ELISA primary screen were subjected to a secondary screen using FACS, as described above.
  • a second batch of several hundreds of hybridomas from fusions mice spleens expressing human antibodies were obtained from a separate round of immunization(s). These were screened for NKG2D-specificity using FACS in the same manner as described in Ex- ample 1. Antibodies from one hybridoma, MS, were selected for recombinant production and further testing.
  • variable regions of the heavy and light chains of the antibodies were identified by PCR and subsequent sequencing of the isolated product, of mRNA from the hybridoma.
  • RNA purification Total RNA was purified using RNeasy from Qiagen according to the manufactures instructions, except that ⁇ -mercaptoethanol was omitted from the procedure. The quality of the RNA was checked by light spectroscopy (260/280 nm, 1.8 ⁇ ratio ⁇ 2.0) and occasionally RNA degradation was evaluated using a bioanalyser.
  • RT-PCR Full length cDNA was synthesised by SMART-RACE (kit from Clonetech). PCR. PCR was performed with the HFII polymerase from Clonetech. The 5' primer
  • VH IgG
  • VL kappa chains
  • VH 3' primers
  • Restriction sites were also present in the 3' primers (BsiWI (VL) and Nhel (VH)).
  • the PCR was performed in duplicate (to check for PCR introduced mutations) for all VH and VL amplifications. If the PCR reaction failed, the VL and VH were amplified using a degenerate 5 ' primer mix from Novagen.
  • PCR product purification The PCR product ( ⁇ 550 bp) was separated on a 1 % agarose gel, excised, purified on GFX columns (from Amersham) and eluted in DNAse free water. Ligation. The PCR products and the expression vector (ampicillin resistance) were cut with appropriate restriction enzymes (VH, EcoRI + Nhel and VL, EcoRI + BsiWI). The ligation of the variable domains into the isotype-dictating vector (lgG4 for NKG2D) was catalyzed by the T4-ligase (Roche). The plasmid used was pTT5 (Durocher et al., Nucleic Acids Res 2002;30(2):e9; Pham et al., Biotechnol Bioeng 2003;84(3):332-42).
  • variable regions of heavy and light chains were inserted into heavy and light chain human lgG4 framework respectively and expressed from two vectors in HEK293 cells at a high level.
  • the antibodies were purified on a protein A column.
  • HEK293/6E cells Antibody expression in HEK293/6E cells.
  • HEK293 cells were passaged in Free- style293 medium from Gibco. On the day of transfection, cells were diluted to a concentration of 1 million cells/ml. For a 30 ml transfection, 15 ⁇ g of heavy-chain vector and 15 ⁇ g of light- chain vector were mixed with 2 ml Opti-MEM and 40 ⁇ l 293fectin (then Freestyle293 medium to a total volume of 30 ml). After 6 days of incubation, cells were pelleted by centrifugation (1000 rpm, 10 min) and the supernatant was harvested for protein A purification.
  • the recombinantly expressed lgG4 variants of the human antibodies was purified on MabSelectTM SuRe protein-A columns. After column application of antibody, the column was washed with 10 column volumes of PBS buffer, and antibody eluted with 100 mM Glycine, 100 mM NaCI buffer, pH 3.0, followed by buffer exchange into PBS buffer using a HighTrapTM Desalting column. All operations were controlled by an Aktaxpress system from GE Healthcare Amersham Biosciences AB.
  • results cDNA sequences encoding 16F16 (lgG4) H chain, 16F16 L chain, 16F31 (lgG4) H chain, and 16F31 L chain are set forth in SEQ ID NOS:3-6, respectively, and respective sequence identifiers of full-length, variable, and CDR amino acid sequences of 16F16 (lgG4), 16F31 (lgG4), MS (lgG4) and 21 F2 (lgG4) are set forth in Table 1.
  • Figure 4 shows the amino acid sequences for 16F16, 16F31 , MS, and 21 F2 of lgG4 isotype, high-lighting variable (bold) and CDR (bold underline) regions.
  • 16F16 VH VH3_21/D3-9/JH4 (SEQ ID NOS:31/32/33, respectively)
  • 16F16 VL VKI_L15/JK2 (SEQ ID NOS:34/35, respectively)
  • VH VH3_20/D3-10/JH6 (SEQ ID NOS:36/(EL)/37, respectively)
  • 16F31 VL VKIII_A27/JK3 (SEQ ID NOS:38/39, respectively)
  • MS VH VH4_59/D3_27_R3/JH3 (SEQ ID NOS:64/(NWG)/65, respectively)
  • Flow cytometry assays - MICA blockade For analysis of blockade of ligand binding, 50000 NKG2D/DAP10-expressing BaF/3 cells were incubated in 100 ⁇ l total (PBS with 2%FBS at pH7.4) with varying amounts of hybridoma supernatant or purified antibody for 1 h at 16 0 C, followed by incubation with mFc-MICA (for human antibodies) or hFc-MICA (for ON72) (1 ⁇ g) for 30 min at 4 0 C. The cells were thereafter washed, and secondary Goat-Anti- mouse IgG-HRP Fey Fragment specific, Jackson (109-036-151) was added for detection of MICA-mFc binding.
  • mFc-MICA for human antibodies
  • hFc-MICA for ON72
  • the cells were then analysed on a B&D FACSArray flow cytometer.
  • the degree of reduction of MICA binding by preincubation was analysed as MFI (mean fluores- cence intensity) of binding with pre-incubation in % of binding of MICA without preincubation.
  • Antibodv IC50 (u ⁇ /ml) Full blockade of 1 u ⁇ MICA-Fc
  • Flow cytometry assay - competition with murine antibodies For analysis of block- ade of commercially available murine anti-hNKG2D antibodies, 50000 NKG2D-expressing cells were incubated in 100 ⁇ l final (PBS with 2%FBS at pH7.4) with hybridoma supernatant or purified and recombinantly expressed antibody (at 0.3 ⁇ g or as indicated) for 1 h at 16 0 C, followed by incubation with a murine anti-hNKG2D antibody (ON72, 149810, 1 D11 or 5C6 (for 1 D1 1 and 5C6, see, e.g., Bauer et a/., Science 1999:285:727-9 and WO02068615); at 0.3 ⁇ g or as indicated) for 30 min at 4 0 C.
  • a murine anti-hNKG2D antibody ON72, 149810, 1 D11 or 5C6 (for 1 D1 1 and 5C6, see, e.g., Bauer e
  • the cells were thereafter washed, and secondary Goat-Anti-mouse IgG-HRP Fey Fragment specific, Jackson (109-036-151) was added for detection of binding of the murine antibody.
  • the cells were analysed on a B&D FACSArray. This setup was also performed by pre-incubation with murine antibody followed by the hybri- doma supernatant or purified human antibody, using secondary Goat-anti-human IgG-HRP Fey Fragment specific (Jackson, 109-036-098) for detection.
  • the degree of reduction of binding by pre-incubation was analysed as MFI of binding with pre-incubation in % of binding without pre-incubation.
  • PBMC Perifieral blood mononuclear cells
  • PMBCs Perifieral blood mononuclear cells
  • NK, CD8+, CD4+ and y ⁇ T cells were isolated from humans, or from Cynomologous or Rhesus monkey. All animal work were performed according to Danish National Research Council guidelines.
  • Each PBMC sample was labelled with a marker for the different cellular subsets (NK, CD8+, CD4+ and y ⁇ T cells, as well as for NKG2D, with either ON72 or recombinantly expressed and purified 16F16.
  • the cells were washed and analysed on a BD FACSDiva (BD Biosource) for staining of subsets of cells for NKG2D with the two antibodies. The MFI of the staining was calculated for the individual antibodies.
  • NKG2D-ligand-driven cytotoxicity assay was developed.
  • a 51Cr-release assay was used, where the target cells were loaded with the radioactive dye and its release measured as a consequence of NK killing of the cell.
  • NKG2D-MICA interaction mediated killing assay Biological assays for measuring NKG2D-ligand mediated killing of target cells are suitable for testing anti-NKG2D antibodies.
  • the NK cell lines NK92 or NKL both kill MICA-transfected BaF/3 cells in an NKG2D-dependent fashion, and can be used as effector cells, killing 51Cr-loaded target cells expressing an NKG2D- ligand (either MICA, MICB or ULBP1-4).
  • NKL cells were incubated for 4h with 51Cr-loaded, MICA-expressing BaF/3 cells in the ratio 10:1 , in the presence or absence of 1 or 5 ⁇ g ON72 or recombinantly expressed 16F16 of lgG4 isotype. After incubation, the supernatant was transferred to micro- titer plates, scintillant added, and the release of 51Cr was measured, as a result of killing of the target cells, in a Topcounter (Wallach). The reduction in release of 51Cr was a measure of inhibition of killing by the added antibody, and the percentage of cells that were killed was calculated.
  • NK-92 cells was incubated with either 51 Cr-labelled MICA- or
  • MS and 21 F2 both inhibited killing of ULBP3-bearing cells by NK-92 cells in a 51Cr-release assay.
  • blocking of cytotoxicity is depicted as % inhibition, with 0 being the two cells incubated together without addition of antibody, with MS being more efficient.
  • maximum inhibition of killing of ligand- (MICA-) bearing cells by NKL cells in a 51 Cr-release assay was obtained at a very low concentration of MS (0.01 ⁇ g/ml) while the highest tested concentration of 16F16 (0.1 ⁇ g/ml) only lead to about 40% inhibition.
  • a summary of IC50 data are provided in Table 5.
  • Antibody EC50 ( ⁇ g/ml)
  • NK cells were incubated with different amounts of MS (0; 0,003; 0.01 ; 0.03; 0.1 ; 0.3; 1 ⁇ g) or 21 F2 (0.1 ⁇ g) in the presence of 10% human serum to mimic the situation in full blood, with the presence of IgGs with higher affinity for the Fc receptors.
  • MS 0,003; 0.01 ; 0.03; 0.1 ; 0.3; 1 ⁇ g
  • 21 F2 0.1 ⁇ g
  • 0.1 ⁇ g /ml MS, ON72, or 21 F2 was added to whole blood containing NK, CD8+, and y ⁇ T cells. After incubation, staining with anti-CD8, anti-CD56 (NK cells), anti- ⁇ , and anti-human antibody identified binding to the various subtypes
  • ON72, 16F16, and 16F31 all induced NKG2D down- modulation in NKG2D-expressing BAF/3 cells.
  • ON72 and 16F31 approximately 55% down-modulation was observed, as compared to about 75% down-modulation using 16F16. This suggests that 16F16 induces down-modulation more effectively, as it has a similar Kd value to that of ON72. Without being limited to theory, this might be due to different binding epitopes. After incubation with MS, there was about 95% reduction in surface NKG2D (Fig- ure 16A).
  • Example 8 Non-depleting lgG4 versions of human antibodies Antibodies cross-linking cells in blood may induce depletion of the antibody-bound cells. However, the affinity of lgG4 antibodies to the activating Fc-receptors is so low compared to that of IgGI that lgG4 antibodies do not lead to depletion. Materials and Methods
  • Biacore 1000 upgrade apparatus Biacore GE Healthcare; Biacore Upgrade CA0396
  • HBS-EP buffer Biacore GE Healthcare; BR-1001-88
  • sensorgrams were analyzed with Biaevaluation 4.1 software.
  • Recombinant MICA-Fc proteins were obtained from R&D systems or were recombinantly produced.
  • Recombinant ULBP-1 , 2, 3, MICB and NKG2D- Fc were purchased from R&D systems.
  • Recombinant NKG2D-Fc proteins were immobilized covalently to carboxyl groups in the dextran layer on a Sensor Chip CM5 (Biacore GE Healthcare; BR- 1000-14).
  • the sensor chip surface was activated with EDC/NHS (N-ethyl- N'-(3-dimethylaminopropyl) carbodiimidehydrochloride and N-hydroxysuccinimide (Biacore GE Healthcare; BR-1000-50)).
  • Proteins were diluted to 10 ⁇ g/ml in coupling buffer (10 mM acetate, pH 5.2) and injected until the appropriate immobilization level was reached (i.e. 500 to 1000 RU). Deactivation of the remaining activated groups was performed using 100 mM ethanolamine pH 8 (Biacore GE Healthcare; BR-1000-50).
  • NKG2D-Fc proteins 500 to 1000 RU
  • Full dose-response curves for binding of antibody to cells expressing NKG2D were performed to analyze binding affinity to the naturally occurring receptor.
  • Figure 3 demonstrates dose-dependent NKG2D binding of recombinantly produced and purified human antibodies 16F16, 16F31 , MS and 21 F2 to NKG2D- and DAP10- expressing BaF/3 cells, as compared to commercially available murine antibodies (ON72 and 149810), using flow cytometry.
  • the EC50 values for binding were as follows:
  • Example 10 Agonist activity of immobilized anti-NKG2D antibodies
  • PBMCs peripheral blood lymphocytes
  • CD3 peripheral blood lymphocytes
  • the stimulation was done under circumstances where NKG2D have been shown to act as a co-stimulatory molecule (Mashoo et al, Immunol. 2005;174;4480-4484), believed to reflect the triggering of NKG2D in the pres- ence of pro-inflammatory cytokines as under chronic inflammatory conditions.
  • PBMCs were stimulated for 3 days with surface bound antibodies, followed by 4 days of IL-2 stimulation, and the proliferation assessed by CFSE dilution in either all lymfocytes, CD8+, or CD4+ T cells.
  • PBMC prolfieration assay PBMCs were purified by gradient centrifugation. Ninety- six-well Maxisorp plates were coated with anti-Fc antibody (Jackson - lmmuno Resarch 115- 006-008), then washed and followed by addition of anti-CD3 (0.1 or 0.3 ng/ml, Bioscience cat#14-0037-82), anti-NKG2D (MS or ON72, 0.2 ⁇ g/ml) and/or anti-CD28 antibody (0.2 ⁇ g/ml, Becton Dickison cat# 348040). Onehundred-and-fifty thousand PBMCs were added to each well, and the cells incubated at 37°C for 3 days.
  • CFSE molecular probes cat# C34554
  • Ten million cells were incubated in 0.5 ml 1 ⁇ M CFSE for 10 min at 37°C, followed by a wash, and 150.000 PBMCs per well in 60-well plates were incubated for 4 days with IL-2 (10 U/ml).
  • IL-2 10 U/ml
  • the cells were stained with anti-CD8 and anti-CD4 antibody and the proliferation measured by CFSE dilution in either all lymphocytes (total presented in Figure 19) or CD8+ or CD4+ T cells (similar results obtained)
  • MS did not significantly co-stimulate proliferation of lymphocytes at either 0.1 or 0.3 ng/ml CD3 stimulation, whereas ON72 resulted in a small but sig- nificant co-stimulation at both CD3 concentrations. See Table 7. In both cases the control, anti-CD28, gave strong co-stimulation, and anti-NKG2D did not add significantly to this. This shows that there is a difference in binding mode of the two antibodies, with immobilized ON72 having detectable agonistic activity, whereas MS is a more pure antagonist.
  • Soluble hNKG2D (residues 89-216 of SEQ ID NO:2) and MS Fab (comprising a light chain corresponding to SEQ ID NO:41 and a heavy chain fragment corresponding to residues 1 -213 of SEQ ID NO: 40) were mixed with a slight molar excess of hNKG2D and the complex was purified on a gel-filtration column. The complex was then concentrated to about 9.5 mg/ml. Crystals were grown with the hanging drop-technique in 17 % PEG3350, 200 mM sodium malonate and 100 mM bis-tris-propane buffer with a pH of 7.5.
  • Crystals were transferred to a cryo-solution containing 75 % of the precipitant solution and 25 % of glycerole. The crystal was allowed to soak for about 15 seconds. The crystal was then flash frozen in liquid N 2 and kept at 100 K during data collection by a cryogenic N 2 gas stream.
  • Crystallographic data were collected, originally to 2.4 A resolution at a Rigaku 007HF rotating anode source and thereafter, using a new crystal, to 1.7 A resolution at beam-line BL911 - 3(2) at MAX-lab, Lund, Sweden. Space group determination, integration and scaling of the data were made in the XDS software package(3). Cell parameters for the synchrotron data were determined to be 82.1 , 54.2, 169.4 A, 90°, 102.62° and 90°, respectively.
  • the structure was originally interpreted in the C2 space group, using the rotating anode data.
  • synchrotron data XDS(3) indicated a non-centric monoclinic space group though and data were integrated in space group P2, later changed to P2 ⁇ A C-centered orthorhombic cell did also score highly.
  • POiNT- LESS software(13) proposed the P2i as the correct space group when testing the synchrotron data.
  • the PHASER software program was used for a new round of molecular replacement, using the preliminary models prepared in the first round of molecular replacement in the C2 space group.
  • MS-Fab effectively blocks MICA binding to both monomers of the hNKG2D dimer.
  • MICA MICA molecule bound to both monomers of the NKG2D dimer(1)
  • MS Fab bound primarily to one of the two monomers, herein denoted "NKG2D monomer unit 1" (Fig. 21 A and 21C).
  • the interactions between MS and NKG2D monomer unit 2 were found to be less specific (e.g., comprise no or fewer hydrogen-bonds), and less important in keeping the MS Fab/NKG2D complex together.
  • the resulting hNKG2D epitope for MS was found to comprise the following residues of hNKG2D (SEQ ID NO: 2): Lys 150, Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, GIn 185, Leu 191 , Lys 197, Tyr 199, GIu 201 , Thr 205, Pro 206, Asn 207 and Thr 208 (Fig. 21A).
  • Lys 150 Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, GIn 185, Leu 191 , Lys 197, Tyr 199, GIu 201 , Thr 205, Pro 206, Asn 207 and Thr 208 (Fig. 21A).
  • NKG2D monomer unit 2 only 5 interactions were found, and only one residue (Tyr 152) was present in both of the crystallographically independent complexes.
  • the MS hNKG2D epitope comprised residues located in the loop just before and the beginning of ⁇ -strand ⁇ 3'(1 ), Lys 150-Tyr 152; in ⁇ 5' and the loop after, Thr 180-GIn 185; in ⁇ 5, Leu 191 ; in ⁇ 6, Lys 197, Tyr 199 and GIu 201 ; and in the loop preceding and in the ⁇ 7 strand, Thr 205-Thr 208.
  • These contact areas agreed very well with what have been reported as the binding site for MICA on hNKG2D (1 ).
  • MICA binds asymmetrically to the symmetric homodimer of NKG2D(10). This is also the case for the MS Fab binding to hNKG2D. Therefore, there will be two possible binding orientations to NKG2D for MS Fab relative to MICA. This is shown in Figure 20 A, B and Figure 22, where it is clearly seen that the MS Fab blocks both possible MICA relative binding orientations.
  • the MS paratope for hNKG2D included residues Tyr 33 and Trp 97 of the MS light (L) chain (SEQ ID NO: 41 , Tables 9-12), and residues GIn 1 , Asp 26, Asp 27, Ser 30, Ser 31 , Tyr 32, Tyr 33, His 50, Ser 52, Tyr 53, Ser 54, Ser 56, Ala 57, Asn 58, Trp 98 and Asp 99 of the heavy (H) chain (SEQ ID NO: 40, Tables 9-12).
  • the hNKG2D epitope, and the residues involved in hydrogen-binding, are also indicated in the amino-acid sequence of hNKG2D in Figure 21A.
  • Example 12 Crystal structure of soluble hNKG2D in complex with hzON72-Fab
  • a soluble hNKG2D fragment (corresponding to residues 81-216 of SEQ ID NO:2) and hzON72 Fab (SEQ ID NO:70 and SEQ ID NO:71 , heavy chain fragment and light chain, respectively) were mixed with a slight molar excess of hNKG2D and the complex was purified on a gel-filtration column. The complex was then concentrated to about 7.5 mg/ml. Crystals were grown with the hanging drop-technique in 1 M LiSO 4 and 100 mM MES buffer pH 6.5. Crystals were transferred to a cryo-solution containing 75 % of the precipitant solu- tion and 25 % of glycerole. The crystal was allowed to soak for about 15 seconds.
  • Crystallographic data to 3.15 A resolution were collected using beam-line BL91 1-5(2) at MAX-lab, Lund, Sweden. Space group determination, integration and scaling of the data were made in the XDS software package(3). Cell parameters were determined to be 65.7, 93.3, 128.9 A, 90°, 93.83° and 90°, respectively. Space group was determined to be P2- I with space for one NKG2D dimer and two hzON72 Fab molecules, in the asymmetric unit.
  • a MICA molecule binds strongly to both monomers of NKG2D(1 ), but two hzON72- Fab molecules instead bound independently to each of the hNKG2D monomers, effectively blocking MICA binding to the NKG2D dimer ( Figures 2OC, 21 B, C and 22).
  • Calculation by the software program AREAIMOL of the CCP4 program suite(7) of the average areas excluded in pair-wise interactions gave for the two crystallographically independent soluble hNKG2D/hzON72-Fab molecular complexes (one hzON72 Fab molecule in complex with one hNKG2D monomer) in the determined crystals structure a total of 791 and 801 A 2 , respectively.
  • the average areas excluded in pair-wise interaction between the soluble hNKG2D monomers and the heavy chains of hzON72-Fab were calculated to be, for the two crystallographically independent complexes, 642 and 631 A 2 , respectively, while for the light chains 208 and 242 A 2 , respectively.
  • the direct contacts between the hNKG2D to hzON72-Fab were identified by running the CONTACTS software of the CCP4 program suite using a cut-off distance of 4.0 A between the hzON72-Fab and hNKG2D molecules.
  • the results for the two independent soluble hNKG2D/hzON72-Fab molecules of the crystal structure are shown in Tables 14-15.
  • the resulting hNKG2D epitope for hzON72 was found to comprise the following residues of hNKG2D (SEQ ID NO: 2): Ser 165, Trp 166, Leu 174, Ser 175, Pro 176, Asn 177, Leu 179, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, Lys 186, Ala 193, Ser 194, Ser 195, Lys 197 and Tyr 199.
  • the hzON72 paratope for hNKG2D included residues Tyr 1 , Lys 92, Thr 93 and
  • the hNKG2D epitope was comprised of residues located in the beginning of ⁇ - strand ⁇ 4(1 ), Ser 165-Trp 166; in the loop before ⁇ 5', Leu 174-Asn 177; in the ⁇ 5' strand and the loop thereafter, Leu 179-Lys 186; the loop before ⁇ 6, Ala 193-Ser 195; and in the ⁇ 6 strand, Lys 197 and Tyr 199.
  • These contact areas agreed very well with what have been re- ported as the binding site for MICA on hNKG2D(1 ) and it was clear that hzON72 antibody can block the MICA binding. This is shown in Figures 2OC, 21 B, C and 22.
  • REMARK REFINEMENT REFINEMENT.
  • REMARK CROSS-VALIDATION METHOD THROUGHOUT REMARK FREE R VALUE TEST SET SELECTION : RANDOM REMARK R VALUE (WORKING + TEST SET) : 0.21870 REMARK R VALUE (WORKING SET) 0.21608 REMARK FREE R VALUE 0.26854 REMARK FREE R VALUE TEST SET SIZE (%) 5.0 REMARK FREE R VALUE TEST SET COUNT 1350 REMARK REMARK FIT IN THE HIGHEST RESOLUTION BIN.
  • REMARK CORRELATION COEFFICIENT FO-FC 0.896 REMARK CORRELATION COEFFICIENT FO-FC FREE : 0.844 REMARK REMARK RMS DEVIATIONS FROM IDEAL VALUES COUNT RMS WEIGHT REMARK BOND LENGTHS REFINED ATOMS (A) 8971 0.012 0.022 REMARK BOND ANGLES REFINED ATOMS (DEGREES) 12204 1 , 516 1.948 REMARK TORSION ANGLES, PERIOD 1 (DEGREES) 1116 7 , 673 5.000 REMARK TORSION ANGLES, PERIOD 2 (DEGREES) 368 37, 729 24.402 REMARK TORSION ANGLES, PERIOD 3 (DEGREES) 1464 21 , 254 15.000 REMARK TORSION ANGLES, PERIOD 4 (DEGREES) 32 19, 705 15.000 REMARK CHIRAL-CENTER RESTRAINTS
  • hNKG2D An isolated human or humanized monoclonal antibody, or antigen-binding fragment thereof, which binds human NKG2D (hNKG2D). 2. The antibody or antigen-binding fragment of the preceding embodiment, which reduces hNKG2D-mediated activation of an hNKG2D-expressing NK or T cell.
  • the antibody or antigen-binding fragment of any preceding embodiment which, when immobilized, does not significantly co-stimulate CD3-triggered proliferation of periph- eral blood mononuclear cells (PBMCs). 7. The antibody or antigen-binding fragment of any preceding embodiment, which, when immobilized, has no significant agonistic effect on hNKG2D-mediated activation of an hNKG2D-expressing NK or T cell.
  • PBMCs periph- eral blood mononuclear cells
  • the antibody of any preceding embodiment which binds to hNKG2D with a KD of 0.1 nM or less.
  • the antibody or antigen-binding fragment of any preceding embodiment which, when added to NKG2D-expressing NK or T cells, cross-links no more than two hNKG2D dimers.
  • the antibody or antigen-binding fragment of any preceding embodiment which binds strongly to only a first hNKG2D monomer in an hNKG2D dimer.
  • the antibody or antigen-binding fragment of the preceding embodiment which, when bound to the first hNKG2D monomer, blocks binding of the antibody or antigen- binding fragment to the second hNKG2D monomer.
  • an antibody comprising a heavy-chain variable region comprising the se- quence of SEQ ID NO:46 and a light-chain variable region comprising the sequence of SEQ ID NO:47.
  • any of the preceding embodiments which binds to the same epitope of NKG2D as a reference antibody
  • the reference antibody is selected from the group consisting of: (a) an antibody comprising a heavy-chain variable region comprising the sequence of SEQ ID NO:11 and a light-chain variable region comprising the sequence of SEQ ID NO:12;
  • an antibody comprising a heavy-chain variable region comprising the se- quence of SEQ ID NO:46 and a light-chain variable region comprising the sequence of SEQ ID NO:47.
  • any of the preceding embodiments which binds to NKG2D with substantially the same KD as a reference antibody
  • the reference antibody is selected from the group consisting of: (a) an antibody comprising a heavy-chain variable region comprising the sequence of SEQ ID NO:11 and a light-chain variable region comprising the sequence of SEQ ID NO:12;
  • an antibody comprising a heavy-chain variable region comprising the sequence of SEQ ID NO:44 and a light-chain variable region comprising the sequence of SEQ ID NO:45; and (d) an antibody comprising a heavy-chain variable region comprising the sequence of SEQ ID NO:46 and a light-chain variable region comprising the sequence of SEQ ID NO:47.
  • the antibody or antigen-binding fragment of the preceding embodiment which binds to an epitope comprising Lys 150, Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, GIn 185, Leu 191 , Lys 197, Tyr 199, GIu 201 , Thr 205, Pro 206, Asn
  • the antibody or antigen-binding fragment of the preceding embodiment which binds to an epitope comprising 5 or more residues selected from Lys 150, Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, GIn 185, Leu 191 , Lys 197, Tyr 199, GIu 201 , Thr 205, Pro 206, Asn 207 and Thr 208 of SEQ ID NO:2.
  • the antibody or antigen-binding fragment of the preceding embodiment which binds to an epitope comprising Lys 150, Ser 151 , Tyr 152, Thr 180, Ne 181 , Ne 182, GIu 183, Met 184, GIn 185, Leu 191 , Lys 197, Tyr 199, GIu 201 , Thr 205, Pro 206, Asn 207 and Thr 208 of SEQ ID NO:2.
  • 23 The antibody or antigen-binding fragment of any of embodiments 16-18, wherein the reference antibody comprises a heavy-chain variable region comprising the sequence of SEQ ID NO:46 and a light-chain variable region comprising the sequence of SEQ ID NO:47.
  • VH4_59, D7_27_R3, and JH3 genes (c) VH4_59, D7_27_R3, and JH3 genes; or (d) VH5_51 , D3_10_R3 and JH4 genes.
  • the antibody or antigen-binding fragment of any preceding embodiment comprising a light-chain variable region that is the product of or derived from a set of human genes comprising (a) VKI_L15 and JK2 genes;
  • VKIII_L6 and JK1 genes (d) VKIII_L6 and JK1 genes. 26.
  • the antibody or antigen-binding fragment of any preceding embodiment comprising a heavy chain variable region that is the product of or derived from a set of human genes comprising VH4_59, D7_27_R3, and JH3 genes, and a light-chain variable region that is the product of or derived from a set of human genes comprising VKIII_A27 and JK1 genes.
  • the antibody or antigen-binding fragment of any preceding embodiment comprising a paratope comprising a residue corresponding to GIn 1 , Asp 26, Asp 27, Ser 30, Ser 31 , Tyr 32, Tyr 33, His 50, Ser 52, Tyr 53, Ser 54, Ser 56, Ala 57, Asn 58, Trp 98 or Asp 99 of SEQ ID NO: 44 or Tyr 33 or Trp 97 of SEQ ID NO:45, or any combination thereof.
  • the antibody or antigen-binding fragment of the preceding embodiment comprising a paratope comprising at least 5 residues selected from the residues corresponding to GIn 1 , Asp 26, Asp 27, Ser 30, Ser 31 , Tyr 32, Tyr 33, His 50, Ser 52, Tyr 53, Ser 54, Ser 56, Ala 57, Asn 58, Trp 98 or Asp 99 of SEQ ID NO: 44 or Tyr 33 or Trp 97 of SEQ ID NO:45, or any combination thereof. 29.
  • the antibody or antigen-binding fragment of the preceding embodiment comprising a paratope comprising residues corresponding to GIn 1 , Asp 26, Asp 27, Ser 30, Ser 31 , Tyr 32, Tyr 33, His 50, Ser 52, Tyr 53, Ser 54, Ser 56, Ala 57, Asn 58, Trp 98 and Asp 99 of SEQ ID NO: 44 and Tyr 33 and Trp 97 of SEQ ID NO:45.
  • a light-chain variable region CDR3 comprising the sequence of SEQ ID NO:53.
  • the antibody or antigen-binding fragment of any preceding embodiment comprising a heavy-chain variable region comprising the sequence of SEQ ID NO:44, and a light- chain variable region comprising the sequence of SEQ ID NO:45.
  • the antibody or antigen-binding fragment of any of embodiments 1-25 comprising a heavy chain variable region that is the product of or derived from a set of human genes comprising VH5_51 , D3_10_R3 and JH4 genes, and a light-chain variable region that is the product of or derived from a set of human genes comprising VKIII_L6 and JK1 genes.
  • the antibody or antigen-binding fragment of the preceding embodiment comprising a heavy-chain variable region comprising the sequence of SEQ ID NO:46, and a light- chain variable region comprising the sequence of SEQ ID NO:47.
  • the antibody or antigen-binding fragment of embodiment 1 comprising:
  • the antibody or antigen-binding fragment of the preceding embodiment comprising a heavy-chain variable region comprising the sequence of SEQ ID NO:1 1 , and a light- chain variable region comprising the sequence of SEQ ID NO:12. 40.
  • the antibody or antigen-binding fragment of embodiment 1 comprising:
  • the antibody or antigen-binding fragment of the preceding embodiment comprising a heavy-chain variable region comprising the sequence of SEQ ID NO: 13, and a light- chain variable region comprising the sequence of SEQ ID NO:14.
  • the antibody or antigen-binding fragment of embodiment 1 comprising: (a) a heavy-chain variable region CDR1 comprising the sequence of SEQ ID NO:48;
  • the antibody or antigen-binding fragment of embodiment 1 comprising a heavy-chain variable region comprising the sequence of SEQ ID NO:44, and a light-chain variable region comprising the sequence of SEQ ID NO:45.
  • the antibody or antigen-binding fragment of the preceding embodiment comprising a heavy-chain variable region comprising the sequence of SEQ ID NO:46, and a light- chain variable region comprising the sequence of SEQ ID NO:47.
  • the antibody or antigen-binding fragment of embodiment 1 comprising
  • the antibody or antigen-binding fragment of any preceding embodiment which is bivalent.
  • the antibody of the preceding embodiment which is an lgG4 antibody.
  • SEQ ID NO:7 and a light-chain sequence comprising the sequence of SEQ ID NO:8.
  • An isolated antibody comprising a heavy-chain sequence comprising the sequence of SEQ ID NO:9 and a light-chain sequence comprising the sequence of SEQ ID NO:10.
  • An isolated antibody comprising a heavy-chain sequence comprising the sequence of SEQ ID NO:40 and a light-chain sequence comprising the sequence of SEQ ID NO:
  • An isolated antibody comprising a heavy-chain sequence comprising the sequence of SEQ ID NO:42 and a light-chain sequence comprising the sequence of SEQ ID NO:43.
  • the antibody of the preceding embodiment which is capable of downmodulating less than 90% of cell-surface-associated NKG2D of an NK cell.
  • 71. An isolated antibody that binds hNKG2D, and which binds to human and cynomol- gous NKG2D-expressing cells with similar affinity.
  • the antibody of the preceding embodiment which has an EC50 for binding to cyno- molgous CD8+ T cells in a PBMC preparation that is at least 50% of its EC50 for binding to human CD8+ T cells in a PBMC preparation.
  • the antibody of the preceding embodiment which has an EC50 for binding to cyno- molgous CD8+ T cells in a PBMC preparation that is at least 65% of its EC50 for binding to human CD8+ T cells in a PBMC preparation.
  • the antibody of any preceding embodiment which has an affinity for human NKG2D of no more than 10 pM.
  • a host cell comprising the expression vector of the preceding embodiment.
  • a host cell producing the antibody or antigen-binding fragment of any of embodiments 1-79.
  • the host cell of any of embodiments 82 and 83 which is a CHO cell.
  • a method of producing an anti-NKG2D antibody or antigen-binding fragment comprising ing culturing the host cell of any of embodiments 82 to 84 under suitable conditions and recovering said antibody or antigen-binding fragment thereof.
  • a method of producing a variant anti-NKG2D antibody, or an antigen-binding fragment thereof, comprising
  • An immunoconjugate comprising the antibody or antigen-binding fragment of any of embodiments 1-79, linked to a therapeutic agent.
  • a multispecific molecule comprising the antibody or antigen-binding fragment of any of embodiments 1-79, linked to a second moiety having a different binding specificity than the antibody.
  • composition comprising the antibody or antigen-binding fragment of any of embodiments 1-79, and a pharmaceutically acceptable carrier.
  • 91. A method for preventing NKG2D-mediated activation of an NKG2D-expressing NK or
  • T cell comprising contacting the NK or T cell with the antibody or antigen-binding fragment of any of embodiments 1-79, wherein the antibody or antigen-binding fragment competes with at least one NKG2D ligand in binding to NKG2D.
  • a method for reducing the amount of NKG2D on the surface of an NKG2D- expressing NK or T cell comprising contacting the NK or T cell with the antibody or antigen-binding fragment of any of embodiments 1 -79, wherein the antibody or antigen-binding fragment competes with at least one NKG2D ligand in binding to human NKG2D.
  • a method for treating an inflammatory or autoimmune disorder comprising adminis- tering the composition of embodiment 90 to a human subject suffering from or at risk for an inflammatory or autoimmune disorder.
  • transplant is a heart trans- plant or a bone marrow transplant.
  • 106 The method of any of embodiments 94 to 105, further comprising administering a second anti-inflammatory agent.
  • the second anti-inflammatory agent is selected from an immunosuppressant, an analgesic, an anti-angiogenic agent, a corticosteroid, a B-cell depletion agent, a B-cell antagonist, a T-cell antagonist, a complement-inhibiting agent, an anti-cytokine agent, and an anti-cytokine receptor agent, and combinations thereof.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Neurology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Dermatology (AREA)
  • Neurosurgery (AREA)
  • Obesity (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Pain & Pain Management (AREA)
  • Transplantation (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
PCT/EP2008/067499 2006-12-21 2008-12-15 Antibodies against human nkg2d and uses thereof WO2009077483A1 (en)

Priority Applications (17)

Application Number Priority Date Filing Date Title
AU2008337517A AU2008337517B2 (en) 2007-12-14 2008-12-15 Antibodies against human NKG2D and uses thereof
DK08861706.3T DK2222706T4 (en) 2007-12-14 2008-12-15 Antibodies that bind to NKG2D and its use
CA2708854A CA2708854C (en) 2007-12-14 2008-12-15 Antibodies against human nkg2d and uses thereof
US12/747,095 US9127064B2 (en) 2006-12-21 2008-12-15 Antibodies against human NKG2D and uses thereof
BRPI0821658A BRPI0821658B8 (pt) 2007-12-14 2008-12-15 anticorpo monoclonal humano ou um fragmento de ligação ao antígeno do mesmo que se liga a hnkg2d e seus usos
JP2010537469A JP5591712B2 (ja) 2007-12-14 2008-12-15 ヒトnkg2dに対する抗体とその使用
EP08861706.3A EP2222706B2 (en) 2007-12-14 2008-12-15 Antibodies against human nkg2d and uses thereof
PL08861706T PL2222706T5 (pl) 2007-12-14 2008-12-15 Przeciwciała przeciwko ludzkiemu NKG2D i ich zastosowania
ES08861706.3T ES2483942T5 (es) 2007-12-14 2008-12-15 Anticuerpos contra NKG2D humano y usos de los mismos
CN200880127171.3A CN101945893B (zh) 2007-12-14 2008-12-15 抗人nkg2d抗体及其用途
RU2010125034/10A RU2563343C2 (ru) 2007-12-14 2008-12-15 Антитела к человеческому nkg2d и их применения
US12/605,187 US7879985B2 (en) 2007-12-14 2009-10-23 Antibodies against human NKG2D and uses thereof
IL205866A IL205866A (en) 2007-12-14 2010-05-20 Human or human antibodies against nkg2d, a method of manufacture and use thereof for drug preparation
ZA2010/03759A ZA201003759B (en) 2007-12-14 2010-05-26 Antibodies against human nkg2d and uses thereof
US14/809,680 US20160024214A1 (en) 2006-12-21 2015-07-27 Antibodies Against Human NKG2D and Uses Thereof
US15/707,393 US10526409B2 (en) 2006-12-21 2017-09-18 Antibodies against human NKG2D and uses thereof
US16/655,646 US20200031939A1 (en) 2007-12-14 2019-10-17 Antibodies Against Human NKG2D and Uses Thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP2007063979 2007-12-14
EPPCT/EP2007/063979 2007-12-14
EP08163163.2 2008-08-28
EP08163163 2008-08-28

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US12/747,095 A-371-Of-International US9127064B2 (en) 2006-12-21 2008-12-15 Antibodies against human NKG2D and uses thereof
US12/605,187 Continuation US7879985B2 (en) 2007-12-14 2009-10-23 Antibodies against human NKG2D and uses thereof
US14/809,680 Continuation US20160024214A1 (en) 2006-12-21 2015-07-27 Antibodies Against Human NKG2D and Uses Thereof

Publications (1)

Publication Number Publication Date
WO2009077483A1 true WO2009077483A1 (en) 2009-06-25

Family

ID=40336450

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/067499 WO2009077483A1 (en) 2006-12-21 2008-12-15 Antibodies against human nkg2d and uses thereof

Country Status (17)

Country Link
US (5) US9127064B2 (ru)
EP (2) EP2222706B2 (ru)
JP (2) JP5591712B2 (ru)
KR (1) KR101615935B1 (ru)
CN (3) CN101945893B (ru)
AU (1) AU2008337517B2 (ru)
BR (1) BRPI0821658B8 (ru)
CA (4) CA3102704A1 (ru)
DK (1) DK2222706T4 (ru)
ES (1) ES2483942T5 (ru)
IL (1) IL205866A (ru)
PL (1) PL2222706T5 (ru)
PT (1) PT2222706E (ru)
RU (1) RU2563343C2 (ru)
TW (1) TWI468174B (ru)
WO (1) WO2009077483A1 (ru)
ZA (1) ZA201003759B (ru)

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010017103A2 (en) * 2008-08-04 2010-02-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Servic Fully human anti-human nkg2d monoclonal antibodies
CN102020717A (zh) * 2010-11-05 2011-04-20 扬州大学 一种增强淋巴细胞靶向杀伤肿瘤的活性因子及其制备方法
WO2011104381A2 (en) 2010-02-26 2011-09-01 Novo Nordisk A/S Stable antibody containing compositions
WO2012000994A1 (en) 2010-06-29 2012-01-05 C.N.R.S. Llt-1 antibodies with new functional properties
US8287861B2 (en) 2008-06-30 2012-10-16 Novo Nordisk A/S Anti-human interleukin-20 antibodies
JP2013502420A (ja) * 2009-08-17 2013-01-24 ザ・ペン・ステート・リサーチ・ファンデーション 心血管疾患、および2型糖尿病などの代謝疾患の治療のためのnkg2d阻害剤の使用
US8454956B2 (en) 2009-08-31 2013-06-04 National Cheng Kung University Methods for treating rheumatoid arthritis and osteoporosis with anti-IL-20 antibodies
US8603470B1 (en) 2012-08-07 2013-12-10 National Cheng Kung University Use of IL-20 antagonists for treating liver diseases
US8613919B1 (en) 2012-08-31 2013-12-24 Bayer Healthcare, Llc High concentration antibody and protein formulations
US20140248289A1 (en) * 2011-08-26 2014-09-04 University Of Cincinnati Methods and Compositions for the Treatment of Respiratory Conditions Via NKG2D Inhibition
US8852588B2 (en) 2012-08-07 2014-10-07 National Cheng Kung University Treating allergic airway disorders using anti-IL-20 receptor antibodies
US9198911B2 (en) 2010-11-02 2015-12-01 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
US9221904B2 (en) 2012-07-19 2015-12-29 National Cheng Kung University Treatment of osteoarthritis using IL-20 antagonists
US9592297B2 (en) 2012-08-31 2017-03-14 Bayer Healthcare Llc Antibody and protein formulations
USRE47150E1 (en) 2010-03-01 2018-12-04 Bayer Healthcare Llc Optimized monoclonal antibodies against tissue factor pathway inhibitor (TFPI)
WO2020127965A1 (en) 2018-12-21 2020-06-25 Onxeo New conjugated nucleic acid molecules and their uses
US10835602B2 (en) 2010-05-28 2020-11-17 Novo Nordisk A/S Stable multi-dose compositions comprising an antibody and a preservative
EP3579866A4 (en) * 2017-02-08 2020-12-09 Dragonfly Therapeutics, Inc. HEAVY CHAIN VARIABLE ANTIBODY DOMAINS WITH NKG2D RECEPTOR AIMING
WO2021009146A1 (en) * 2019-07-15 2021-01-21 F. Hoffmann-La Roche Ag Antibodies binding to nkg2d
EP3661554A4 (en) * 2017-07-31 2021-04-21 Dragonfly Therapeutics, Inc. PROTEINS BINDING TO NKG2D, CD16 AND FLT3
WO2021113853A1 (en) * 2019-12-05 2021-06-10 Vycellix, Inc. Modulators of the immune escape mechanism for universal cell therapy
US11084863B2 (en) 2017-06-30 2021-08-10 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains
EP3749346A4 (en) * 2018-02-08 2021-12-15 Dragonfly Therapeutics, Inc. VARIABLE DOMAINS OF ANTIBODIES TARGETING THE NKG2D RECEIVER
WO2021255223A1 (en) 2020-06-19 2021-12-23 Onxeo New conjugated nucleic acid molecules and their uses
WO2022074206A1 (en) 2020-10-08 2022-04-14 Affimed Gmbh Trispecific binders
US11542332B2 (en) 2016-03-26 2023-01-03 Bioatla, Inc. Anti-CTLA4 antibodies, antibody fragments, their immunoconjugates and uses thereof
WO2023007023A1 (en) 2021-07-30 2023-02-02 Affimed Gmbh Duplexbodies
WO2023014863A1 (en) 2021-08-05 2023-02-09 Go Therapeutics, Inc. Anti-glyco-muc4 antibodies and their uses
WO2023034571A1 (en) 2021-09-03 2023-03-09 Go Therapeutics, Inc. Anti-glyco-lamp1 antibodies and their uses
WO2023034569A1 (en) 2021-09-03 2023-03-09 Go Therapeutics, Inc. Anti-glyco-cmet antibodies and their uses
US11618776B2 (en) 2018-12-20 2023-04-04 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15/IL-15RA and NKG2D antigen binding domains
WO2023111203A1 (en) 2021-12-16 2023-06-22 Onxeo New conjugated nucleic acid molecules and their uses
US11834506B2 (en) 2017-02-08 2023-12-05 Dragonfly Therapeutics, Inc. Multi-specific binding proteins that bind NKG2D, CD16, and a tumor-associated antigen for activation of natural killer cells and therapeutic uses thereof to treat cancer
US11884732B2 (en) 2017-02-20 2024-01-30 Dragonfly Therapeutics, Inc. Proteins binding HER2, NKG2D and CD16
WO2024056861A1 (en) 2022-09-15 2024-03-21 Avidicure Ip B.V. Multispecific antigen binding proteins for stimulating nk cells and use thereof

Families Citing this family (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL2222706T5 (pl) * 2007-12-14 2017-09-29 Novo Nordisk As Przeciwciała przeciwko ludzkiemu NKG2D i ich zastosowania
US9796788B2 (en) 2010-02-08 2017-10-24 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
US20130045492A1 (en) 2010-02-08 2013-02-21 Regeneron Pharmaceuticals, Inc. Methods For Making Fully Human Bispecific Antibodies Using A Common Light Chain
PT2501817E (pt) 2010-02-08 2015-11-04 Regeneron Pharma Rato de cadeia leve comum
NZ611269A (en) * 2010-11-19 2015-03-27 Eisai R&D Man Co Ltd Neutralizing anti-ccl20 antibodies
ES2758051T3 (es) 2011-08-05 2020-05-04 Regeneron Pharma Ratones con cadena ligera universal humanizada
US10040853B2 (en) * 2011-09-09 2018-08-07 Fred Hutchinson Cancer Research Center Methods and compositions involving NKG2D inhibitors and cancer
SI2883449T1 (en) 2012-03-16 2018-05-31 Regeneron Pharmaceuticals, Inc. Light-chain modified antibodies with histidine and genetically modified rodents for their production
SG10202107388VA (en) 2012-03-16 2021-08-30 Regeneron Pharma Non-human animals expressing ph-sensitive immunoglobulin sequences
PL3578201T3 (pl) * 2012-06-28 2023-09-11 University Of Central Florida Research Foundation Incorporated Metody i kompozycje dla naturalnych komórek cytotoksycznych
EP3016973A1 (en) * 2013-07-05 2016-05-11 INSERM - Institut National de la Santé et de la Recherche Médicale Novel alternative splice transcripts for mhc class i related chain alpha (mica) and uses thereof
DE102014001481A1 (de) 2013-10-28 2015-04-30 Euroimmun Medizinische Labordiagnostika Ag Verbesserte Vorrichtung und Verfahren für Reaktionen zwischen einer festen und einer flüssigen Phase
SG11201607203XA (en) 2014-03-21 2016-09-29 Regeneron Pharma Non-human animals that make single domain binding proteins
US10793613B2 (en) 2014-12-15 2020-10-06 Washington University Compositions and methods for targeted cytokine delivery
EP3271403A1 (en) 2015-03-19 2018-01-24 Regeneron Pharmaceuticals, Inc. Non-human animals that select for light chain variable regions that bind antigen
WO2016154585A1 (en) 2015-03-26 2016-09-29 Charles Sentman Anti-mica antigen binding fragments, fusion molecules, cells which express and methods of using
CN109071679B (zh) * 2016-02-05 2023-07-28 华盛顿大学 用于靶向的细胞因子递送的组合物和方法
AT518590A3 (de) 2016-04-29 2022-09-15 Mosbach Klaus Speicherung von binärkodes in molekular geprägten polymeren
KR101926166B1 (ko) * 2016-05-24 2018-12-06 재단법인 아산사회복지재단 수용체 시너지 활성을 이용한 nk 세포의 활성도 검사 방법 및 이를 이용한 nk 세포의 활성도가 관련된 질환의 진단 방법
GB201613167D0 (en) 2016-07-29 2016-09-14 Univ Southampton Cancer and b-cell related disease therapy
EP3500293A4 (en) * 2016-08-19 2020-08-19 Janssen Biotech, Inc. METHODS OF TREATING CROHN'S DISEASE WITH AN ANTI-NKG2D ANTIBODY
US10531275B2 (en) * 2016-12-12 2020-01-07 Commscope Technologies Llc Cluster neighbor discovery in centralized radio access network using transport network layer (TNL) address discovery
EP3579878A4 (en) * 2017-02-10 2020-11-18 Dragonfly Therapeutics, Inc. PROTEINS BINDING PSMA, NKG2D AND CD16
SG11201907648XA (en) * 2017-02-20 2019-09-27 Dragonfly Therapeutics Inc Proteins binding gd2, nkg2d and cd16
JP2020510646A (ja) * 2017-02-20 2020-04-09 ドラゴンフライ セラピューティクス, インコーポレイテッド Cd33、nkg2d及びcd16と結合するタンパク質
US20200002436A1 (en) 2017-02-20 2020-01-02 Dragonfly Therapeutics, Inc. Proteins binding cd123, nkg2d and cd16
WO2018157147A1 (en) 2017-02-27 2018-08-30 Dragonfly Therapeutics, Inc. Multispecific binding proteins targeting caix, ano1, mesothelin,trop2, cea, or claudin-18.2
WO2018165913A1 (zh) 2017-03-15 2018-09-20 南京凯地生物科技有限公司 靶向nkg2dl的特异性嵌合抗原受体及其car-t细胞以及它们的应用
JP2020522473A (ja) * 2017-05-23 2020-07-30 ドラゴンフライ セラピューティクス, インコーポレイテッド Nkg2d、cd16、および腫瘍関連抗原に結合するタンパク質
AU2018271872A1 (en) * 2017-05-23 2019-12-12 Dragonfly Therapeutics, Inc. A protein binding NKG2D, CD16 and ROR1 or ROR2
US20200216544A1 (en) 2017-08-16 2020-07-09 Dragonfly Therapeutics, Inc. Proteins binding nkg2d, cd16, and egfr, hla-e, ccr4, or pd-l1
SG11201913968VA (en) * 2017-08-23 2020-01-30 Dragonfly Therapeutics Inc Proteins binding nkg2d, cd16 and a tumor-associated antigen
US20200277384A1 (en) * 2017-09-14 2020-09-03 Dragonfly Therapeutics, Inc. Proteins binding nkg2d, cd16, and c-type lectin-like molecule-1 (cll-1)
EA202091888A1 (ru) * 2018-08-08 2020-10-23 Драгонфлай Терапьютикс, Инк. Вариабельные домены антител, нацеленные на рецептор nkg2d
EP3836951A1 (en) 2018-08-13 2021-06-23 Signablok, Inc. Peptides and compositions for targeted treatment and imaging
WO2021030588A1 (en) * 2019-08-13 2021-02-18 Courier Therapeutics, Inc. Orthopoxvirus major histocompatibility complex (mhc) class-i like protein (omcp) for treatment of autoimmune disease
WO2022053652A1 (en) 2020-09-11 2022-03-17 Engmab Sàrl Combination therapy for cancer
EP4251202A1 (en) * 2020-11-30 2023-10-04 Merck Sharp & Dohme LLC Arginase 1 binders for inhibiting arginase 1 activity
WO2023281120A1 (en) 2021-07-09 2023-01-12 Luxembourg Institute Of Health (Lih) Dimeric protein complexes and uses thereof
CN113502267A (zh) * 2021-08-24 2021-10-15 羽铂精制生物技术(成都)有限公司 一种用于外周血中nk细胞扩增的培养基及方法

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050158307A1 (en) * 2003-04-22 2005-07-21 Fred Hutchinson Cancer Research Center Methods and compositions for treating autoimmune diseases or conditions
US20060280755A1 (en) * 2004-04-05 2006-12-14 The Regents Of The University Of California Modulation of NKG2D
WO2007005874A2 (en) * 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)

Family Cites Families (111)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4318980A (en) 1978-04-10 1982-03-09 Miles Laboratories, Inc. Heterogenous specific binding assay employing a cycling reactant as label
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4634665A (en) 1980-02-25 1987-01-06 The Trustees Of Columbia University In The City Of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US5179017A (en) 1980-02-25 1993-01-12 The Trustees Of Columbia University In The City Of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
JPS5896026A (ja) 1981-10-30 1983-06-07 Nippon Chemiphar Co Ltd 新規ウロキナ−ゼ誘導体およびその製造法ならびにそれを含有する血栓溶解剤
US4609546A (en) 1982-06-24 1986-09-02 Japan Chemical Research Co., Ltd. Long-acting composition
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0154316B1 (en) 1984-03-06 1989-09-13 Takeda Chemical Industries, Ltd. Chemically modified lymphokine and production thereof
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US4766106A (en) 1985-06-26 1988-08-23 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polymer conjugation
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
US4881175A (en) 1986-09-02 1989-11-14 Genex Corporation Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
US5260203A (en) 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
JP3101690B2 (ja) 1987-03-18 2000-10-23 エス・ビィ・2・インコーポレイテッド 変性抗体の、または変性抗体に関する改良
US5013653A (en) 1987-03-20 1991-05-07 Creative Biomolecules, Inc. Product and process for introduction of a hinge region into a fusion protein to facilitate cleavage
AU612370B2 (en) 1987-05-21 1991-07-11 Micromet Ag Targeted multifunctional proteins
US5091513A (en) 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5132405A (en) 1987-05-21 1992-07-21 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
GB8717430D0 (en) 1987-07-23 1987-08-26 Celltech Ltd Recombinant dna product
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
GB8809129D0 (en) 1988-04-18 1988-05-18 Celltech Ltd Recombinant dna methods vectors and host cells
US5476996A (en) 1988-06-14 1995-12-19 Lidak Pharmaceuticals Human immune system in non-human animal
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
JP2989002B2 (ja) 1988-12-22 1999-12-13 キリン―アムジエン・インコーポレーテツド 化学修飾顆粒球コロニー刺激因子
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US6750325B1 (en) 1989-12-21 2004-06-15 Celltech R&D Limited CD3 specific recombinant antibody
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
EP1690934A3 (en) 1990-01-12 2008-07-30 Abgenix, Inc. Generation of xenogeneic antibodies
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
GB9014932D0 (en) 1990-07-05 1990-08-22 Celltech Ltd Recombinant dna product and method
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5165424A (en) 1990-08-09 1992-11-24 Silverman Harvey N Method and system for whitening teeth
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
CA2089661C (en) 1990-08-29 2007-04-03 Nils Lonberg Transgenic non-human animals capable of producing heterologous antibodies
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US6255458B1 (en) 1990-08-29 2001-07-03 Genpharm International High affinity human antibodies and human antibodies against digoxin
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US6300129B1 (en) 1990-08-29 2001-10-09 Genpharm International Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
DE69233254T2 (de) 1991-06-14 2004-09-16 Genentech, Inc., South San Francisco Humanisierter Heregulin Antikörper
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
US5885793A (en) 1991-12-02 1999-03-23 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
JPH05244982A (ja) 1991-12-06 1993-09-24 Sumitomo Chem Co Ltd 擬人化b−b10
US5869619A (en) 1991-12-13 1999-02-09 Xoma Corporation Modified antibody variable domains
JP4157160B2 (ja) 1991-12-13 2008-09-24 ゾーマ テクノロジー リミテッド 改変抗体可変領域の調製のための方法
EP0746609A4 (en) 1991-12-17 1997-12-17 Genpharm Int NON-HUMAN TRANSGENIC ANIMALS CAPABLE OF PRODUCING HETEROLOGOUS ANTIBODIES
US5777085A (en) 1991-12-20 1998-07-07 Protein Design Labs, Inc. Humanized antibodies reactive with GPIIB/IIIA
DE69333807T2 (de) 1992-02-06 2006-02-02 Chiron Corp., Emeryville Marker für krebs und biosynthetisches bindeprotein dafür
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
CA2118508A1 (en) 1992-04-24 1993-11-11 Elizabeth S. Ward Recombinant production of immunoglobulin-like domains in prokaryotic cells
AU6819494A (en) 1993-04-26 1994-11-21 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
JPH08511420A (ja) 1993-06-16 1996-12-03 セルテック・セラピューテイクス・リミテッド 抗 体
US5641870A (en) 1995-04-20 1997-06-24 Genentech, Inc. Low pH hydrophobic interaction chromatography for antibody purification
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US5697151A (en) 1995-08-07 1997-12-16 General Electric Company Method for repairing partitions of a turbine diaphragm
AU2527397A (en) 1996-03-13 1997-10-01 Protein Design Labs, Inc. Fas ligand fusion proteins and their uses
EP0954340B1 (en) 1996-05-03 2007-06-27 Immunomedics, Inc. Targeted combination immunotherapy of cancer
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
EP0981548A4 (en) 1997-04-30 2005-11-23 Enzon Inc SINGLE CHAIN PROTEINS FIXING ANTIGENS CAPABLE OF GLYCOSYLATION, PRODUCTION AND USES THEREOF
US20020062010A1 (en) 1997-05-02 2002-05-23 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
WO1998052976A1 (en) 1997-05-21 1998-11-26 Biovation Limited Method for the production of non-immunogenic proteins
US20030103984A1 (en) 1998-05-04 2003-06-05 Heinz Kohler Fusion proteins of biologically active peptides and antibodies
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
PT1071700E (pt) 1998-04-20 2010-04-23 Glycart Biotechnology Ag Modificação por glicosilação de anticorpos para melhorar a citotoxicidade celular dependente de anticorpos
PL209786B1 (pl) 1999-01-15 2011-10-31 Genentech Inc Przeciwciało zawierające wariant regionu Fc ludzkiej IgG1, przeciwciało wiążące czynnik wzrostu śródbłonka naczyń oraz immunoadhezyna
ES2569919T3 (es) 1999-04-09 2016-05-13 Kyowa Hakko Kirin Co., Ltd. Procedimiento para controlar la actividad de una molécula inmunofuncional
EP1210372B1 (en) 1999-07-29 2008-01-23 Medarex, Inc. Human monoclonal antibodies to her2/neu
CN1371416B (zh) 1999-08-24 2012-10-10 梅达里克斯公司 人ctla-4抗体及其应用
US6414218B1 (en) 2000-01-18 2002-07-02 The General Hospital Corporation Mouse model for rheumatoid arthritis
JP2004500108A (ja) 2000-03-24 2004-01-08 マイクロメット アーゲー Nkg2dレセプター複合体のエピトープへの結合部位を含む多機能ポリペプチド
TWI313299B (en) 2000-11-30 2009-08-11 Medarex Inc Transgenic transchromosomal rodents for making human antibodies
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US20040031066A9 (en) 2001-01-18 2004-02-12 Faustman Denise L. Mouse model for rheumatoid arthritis
US20040115198A1 (en) 2001-02-28 2004-06-17 Fred Hutchinson Cancer Research Center Activation of lymphocyte populations expressing NKG2D using anti-NKG2D antibodies and ligand derivatives
WO2002068615A2 (en) 2001-02-28 2002-09-06 Fred Hutchinson Cancer Research Center, Inc. Activation of lymphocyte populations expressing nkg2d using anti-nkg2d antibodies and ligand derivatives
WO2002092780A2 (en) 2001-05-17 2002-11-21 Diversa Corporation Novel antigen binding molecules for therapeutic, diagnostic, prophylactic, enzymatic, industrial, and agricultural applications, and methods for generating and screening thereof
BR0213761A (pt) 2001-10-25 2005-04-12 Genentech Inc Composições, preparação farmacêutica, artigo industrializado, método de tratamento de mamìferos, célula hospedeira, método para a produção de uma glicoproteìna e uso da composição
WO2003074679A2 (en) 2002-03-01 2003-09-12 Xencor Antibody optimization
CA2483343A1 (en) 2002-04-22 2003-10-30 Fred Hutchinson Cancer Research Center Soluble mic polypeptides as markers for diagnosis, prognosis and treatment of cancer and autoimmune diseases or conditions
ATE483472T1 (de) 2002-05-30 2010-10-15 Macrogenics Inc Cd16a bindungsproteine und verwendung zur behandlung von immunkrankheiten
EP1413316A1 (en) 2002-09-27 2004-04-28 Bruno Robert Bifunctional conjugates or fusion proteins
DE10261223A1 (de) 2002-12-20 2004-07-08 MedInnova Gesellschaft für medizinische Innovationen aus akademischer Forschung mbH Steigerung der Immunantwort durch Substanzen, welche die Funktion von Natürlichen Killerzellen beeinflussen
JP5642328B2 (ja) * 2003-07-24 2014-12-17 イナート・ファルマ・ソシエテ・アノニムInnate Pharma Pharma S.A. Nk細胞増強化合物を使用する治療抗体の有効性を増大させる方法および組成物
WO2005040219A1 (en) 2003-10-28 2005-05-06 Novo Nordisk A/S Laminin-5 gamma2-binding peptides, related compositions, and use thereof
PL1732588T3 (pl) 2004-04-05 2009-12-31 Univ California Modulowanie NKG2D
WO2005115517A2 (en) 2004-04-28 2005-12-08 Joslin Diabetes Center, Inc. Methods of treating diabetes
DE102004042894A1 (de) 2004-08-30 2006-03-02 Eberhard-Karls-Universität Tübingen Verwendung von Blockern der NKG2D-Rezeptor-/NKG2D-Liganden-Interaktion bei Autoimmunerkrankungen
CN1291998C (zh) 2004-11-26 2006-12-27 中国科学技术大学 Nkg2d受体的配体及其应用
WO2007042573A2 (en) 2005-10-14 2007-04-19 Innate Pharma Compositions and methods for treating proliferative disorders
PL2222706T5 (pl) * 2007-12-14 2017-09-29 Novo Nordisk As Przeciwciała przeciwko ludzkiemu NKG2D i ich zastosowania

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050158307A1 (en) * 2003-04-22 2005-07-21 Fred Hutchinson Cancer Research Center Methods and compositions for treating autoimmune diseases or conditions
US20060280755A1 (en) * 2004-04-05 2006-12-14 The Regents Of The University Of California Modulation of NKG2D
WO2007005874A2 (en) * 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BAUER S ET AL: "Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA", SCIENCE, AMERICAN ASSOCIATION FOR THE ADVANCEMENT OF SCIENCE, US, WASHINGTON, DC, vol. 285, no. 5428, 30 July 1999 (1999-07-30), pages 727 - 729, XP002177592, ISSN: 0036-8075 *

Cited By (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8287861B2 (en) 2008-06-30 2012-10-16 Novo Nordisk A/S Anti-human interleukin-20 antibodies
WO2010017103A3 (en) * 2008-08-04 2010-04-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Servic Fully human anti-human nkg2d monoclonal antibodies
US9273136B2 (en) 2008-08-04 2016-03-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Fully human anti-human NKG2D monoclonal antibodies
WO2010017103A2 (en) * 2008-08-04 2010-02-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Servic Fully human anti-human nkg2d monoclonal antibodies
JP2016014030A (ja) * 2009-08-17 2016-01-28 ザ・ペン・ステート・リサーチ・ファンデーション 心血管疾患、および2型糖尿病などの代謝疾患の治療のためのnkg2d阻害剤の使用
JP2013502420A (ja) * 2009-08-17 2013-01-24 ザ・ペン・ステート・リサーチ・ファンデーション 心血管疾患、および2型糖尿病などの代謝疾患の治療のためのnkg2d阻害剤の使用
US8454956B2 (en) 2009-08-31 2013-06-04 National Cheng Kung University Methods for treating rheumatoid arthritis and osteoporosis with anti-IL-20 antibodies
JP2013520476A (ja) * 2010-02-26 2013-06-06 ノヴォ ノルディスク アー/エス 安定な抗体含有組成物
US9795674B2 (en) 2010-02-26 2017-10-24 Novo Nordisk A/S Stable antibody containing compositions
EP3708190A1 (en) 2010-02-26 2020-09-16 Novo Nordisk A/S Stable antibody containing compositions
EP3216462A2 (en) 2010-02-26 2017-09-13 Novo Nordisk A/S Stable antibody containing compositions
US10709782B2 (en) 2010-02-26 2020-07-14 Novo Nordisk A/S Stable antibody containing compositions
JP2016047858A (ja) * 2010-02-26 2016-04-07 ノヴォ ノルディスク アー/エス 安定な抗体含有組成物
EP3409289A2 (en) 2010-02-26 2018-12-05 Novo Nordisk A/S Stable antibody containing compositions
WO2011104381A2 (en) 2010-02-26 2011-09-01 Novo Nordisk A/S Stable antibody containing compositions
AU2011219715B2 (en) * 2010-02-26 2014-12-11 Novo Nordisk A/S Stable antibody containing compositions
WO2011104381A3 (en) * 2010-02-26 2012-10-11 Novo Nordisk A/S Stable antibody containing compositions
USRE47150E1 (en) 2010-03-01 2018-12-04 Bayer Healthcare Llc Optimized monoclonal antibodies against tissue factor pathway inhibitor (TFPI)
US10835602B2 (en) 2010-05-28 2020-11-17 Novo Nordisk A/S Stable multi-dose compositions comprising an antibody and a preservative
WO2012000994A1 (en) 2010-06-29 2012-01-05 C.N.R.S. Llt-1 antibodies with new functional properties
EP3443981A1 (en) 2010-06-29 2019-02-20 Centre National de la Recherche Scientifique (CNRS) Llt-1 antibodies with new functional properties
US9198911B2 (en) 2010-11-02 2015-12-01 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
US10265258B2 (en) 2010-11-02 2019-04-23 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
US9730877B2 (en) 2010-11-02 2017-08-15 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
US9737469B2 (en) 2010-11-02 2017-08-22 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
US11806555B2 (en) 2010-11-02 2023-11-07 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
US9763866B2 (en) 2010-11-02 2017-09-19 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
US11298570B2 (en) 2010-11-02 2022-04-12 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
US10994157B2 (en) 2010-11-02 2021-05-04 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
US9895301B2 (en) 2010-11-02 2018-02-20 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
CN102020717B (zh) * 2010-11-05 2012-09-26 扬州大学 一种增强淋巴细胞靶向杀伤肿瘤的活性因子及其制备方法
CN102020717A (zh) * 2010-11-05 2011-04-20 扬州大学 一种增强淋巴细胞靶向杀伤肿瘤的活性因子及其制备方法
US20140248289A1 (en) * 2011-08-26 2014-09-04 University Of Cincinnati Methods and Compositions for the Treatment of Respiratory Conditions Via NKG2D Inhibition
US9221904B2 (en) 2012-07-19 2015-12-29 National Cheng Kung University Treatment of osteoarthritis using IL-20 antagonists
US9365652B2 (en) 2012-08-07 2016-06-14 National Cheng Kung University Use of IL-20 antagonists for treating liver diseases
US8852588B2 (en) 2012-08-07 2014-10-07 National Cheng Kung University Treating allergic airway disorders using anti-IL-20 receptor antibodies
US8603470B1 (en) 2012-08-07 2013-12-10 National Cheng Kung University Use of IL-20 antagonists for treating liver diseases
US9849181B2 (en) 2012-08-31 2017-12-26 Bayer Healthcare Llc High concentration antibody and protein formulations
US8613919B1 (en) 2012-08-31 2013-12-24 Bayer Healthcare, Llc High concentration antibody and protein formulations
US9592297B2 (en) 2012-08-31 2017-03-14 Bayer Healthcare Llc Antibody and protein formulations
US11542332B2 (en) 2016-03-26 2023-01-03 Bioatla, Inc. Anti-CTLA4 antibodies, antibody fragments, their immunoconjugates and uses thereof
US11834506B2 (en) 2017-02-08 2023-12-05 Dragonfly Therapeutics, Inc. Multi-specific binding proteins that bind NKG2D, CD16, and a tumor-associated antigen for activation of natural killer cells and therapeutic uses thereof to treat cancer
EP3579866A4 (en) * 2017-02-08 2020-12-09 Dragonfly Therapeutics, Inc. HEAVY CHAIN VARIABLE ANTIBODY DOMAINS WITH NKG2D RECEPTOR AIMING
US11884732B2 (en) 2017-02-20 2024-01-30 Dragonfly Therapeutics, Inc. Proteins binding HER2, NKG2D and CD16
US11084863B2 (en) 2017-06-30 2021-08-10 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains
EP3661554A4 (en) * 2017-07-31 2021-04-21 Dragonfly Therapeutics, Inc. PROTEINS BINDING TO NKG2D, CD16 AND FLT3
EP3749346A4 (en) * 2018-02-08 2021-12-15 Dragonfly Therapeutics, Inc. VARIABLE DOMAINS OF ANTIBODIES TARGETING THE NKG2D RECEIVER
US11939384B1 (en) 2018-02-08 2024-03-26 Dragonfly Therapeutics, Inc. Antibody variable domains targeting the NKG2D receptor
US11884733B2 (en) 2018-02-08 2024-01-30 Dragonfly Therapeutics, Inc. Antibody variable domains targeting the NKG2D receptor
US11618776B2 (en) 2018-12-20 2023-04-04 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15/IL-15RA and NKG2D antigen binding domains
WO2020127965A1 (en) 2018-12-21 2020-06-25 Onxeo New conjugated nucleic acid molecules and their uses
US11827711B2 (en) 2019-07-15 2023-11-28 Hoffmann-La Roche Inc. Antibodies binding to NKG2D
WO2021009146A1 (en) * 2019-07-15 2021-01-21 F. Hoffmann-La Roche Ag Antibodies binding to nkg2d
WO2021113853A1 (en) * 2019-12-05 2021-06-10 Vycellix, Inc. Modulators of the immune escape mechanism for universal cell therapy
WO2021255223A1 (en) 2020-06-19 2021-12-23 Onxeo New conjugated nucleic acid molecules and their uses
WO2022074206A1 (en) 2020-10-08 2022-04-14 Affimed Gmbh Trispecific binders
WO2023007023A1 (en) 2021-07-30 2023-02-02 Affimed Gmbh Duplexbodies
WO2023014863A1 (en) 2021-08-05 2023-02-09 Go Therapeutics, Inc. Anti-glyco-muc4 antibodies and their uses
WO2023034569A1 (en) 2021-09-03 2023-03-09 Go Therapeutics, Inc. Anti-glyco-cmet antibodies and their uses
WO2023034571A1 (en) 2021-09-03 2023-03-09 Go Therapeutics, Inc. Anti-glyco-lamp1 antibodies and their uses
WO2023111203A1 (en) 2021-12-16 2023-06-22 Onxeo New conjugated nucleic acid molecules and their uses
WO2024056861A1 (en) 2022-09-15 2024-03-21 Avidicure Ip B.V. Multispecific antigen binding proteins for stimulating nk cells and use thereof
WO2024056862A1 (en) 2022-09-15 2024-03-21 Avidicure Ip B.V. Multispecific antigen binding proteins for tumor-targeting of nk cells and use thereof

Also Published As

Publication number Publication date
EP2222706A1 (en) 2010-09-01
ES2483942T5 (es) 2017-02-06
JP2011506406A (ja) 2011-03-03
BRPI0821658B1 (pt) 2020-04-22
DK2222706T4 (en) 2016-11-21
RU2010125034A (ru) 2012-01-20
CN105001333A (zh) 2015-10-28
BRPI0821658B8 (pt) 2021-05-25
IL205866A (en) 2015-03-31
US20160024214A1 (en) 2016-01-28
CN105001333B (zh) 2019-05-17
BRPI0821658A2 (pt) 2015-07-07
TWI468174B (zh) 2015-01-11
EP2769993A1 (en) 2014-08-27
PT2222706E (pt) 2014-07-28
US10526409B2 (en) 2020-01-07
RU2563343C2 (ru) 2015-09-20
CN101945893A (zh) 2011-01-12
AU2008337517B2 (en) 2014-06-26
KR101615935B1 (ko) 2016-04-28
JP2015013861A (ja) 2015-01-22
CA3102704A1 (en) 2009-06-25
EP2222706B1 (en) 2014-04-23
US7879985B2 (en) 2011-02-01
CA3102679A1 (en) 2009-06-25
ES2483942T3 (es) 2014-08-08
PL2222706T3 (ru) 2014-09-30
CA2982321C (en) 2021-01-05
JP5591712B2 (ja) 2014-09-17
US20180105594A1 (en) 2018-04-19
AU2008337517A1 (en) 2009-06-25
TW200940089A (en) 2009-10-01
JP5933646B2 (ja) 2016-06-15
US20100272718A1 (en) 2010-10-28
CA2708854A1 (en) 2009-06-25
DK2222706T3 (da) 2014-07-07
US20100056764A1 (en) 2010-03-04
US20200031939A1 (en) 2020-01-30
EP2222706B2 (en) 2016-08-10
US9127064B2 (en) 2015-09-08
IL205866A0 (en) 2010-11-30
KR20100098424A (ko) 2010-09-06
ZA201003759B (en) 2011-04-28
CN101945893B (zh) 2015-02-25
PL2222706T5 (pl) 2017-09-29
CA2708854C (en) 2017-11-28
CN110698561A (zh) 2020-01-17
CA2982321A1 (en) 2009-06-25

Similar Documents

Publication Publication Date Title
US10526409B2 (en) Antibodies against human NKG2D and uses thereof
US8287861B2 (en) Anti-human interleukin-20 antibodies
US20140120105A1 (en) Therapeutic targeting of ficolin-3

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880127171.3

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08861706

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2008861706

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 205866

Country of ref document: IL

Ref document number: 2008337517

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 3702/DELNP/2010

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: MX/A/2010/006330

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2008337517

Country of ref document: AU

Date of ref document: 20081215

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2708854

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2010537469

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20107014371

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12747095

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2010125034

Country of ref document: RU

ENP Entry into the national phase

Ref document number: PI0821658

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20100611